Current Genetic Models for Prediction of Primary Myelofibrosis

LB Polushkina1, VA Shuvaev1, MS Fominykh1, YuA Krivolapov2, EA Belyakova2, ZP Asaulenko2, EV Motyko1, LS Martynenko1, MP Bakai1, NYu Tsybakova1, SV Voloshin1,3, SS Bessmeltsev1, AV Chechetkin1, IS Martynkevich1

1 Russian Research Institute of Hematology and Transfusiology, 16 2-ya Sovetskaya str., Saint Petersburg, Russian Federation, 191024

2 II Mechnikov North-Western State Medical University, 41 Kirochnaya str., Saint Petersburg, Russian Federation, 191015

3 SM Kirov Military Medical Academy, 6 Akademika Lebedeva str., Saint Petersburg, Russian Federation, 194044

For correspondence: Lyubov Borisovna Polushkina, PhD in Biology, 16 2-ya Sovetskaya str., Saint Petersburg, Russian Federation, 191024; e-mail: polushkina.lb@gmail.com

For citation: Polushkina LB, Shuvaev VA, Fominykh MS, et al. Current Genetic Models for Prediction of Primary Myelofibrosis. Clinical oncohematology. 2019;12(4):391–7 (In Russ).

DOI: 10.21320/2500-2139-2019-12-4-391-397


ABSTRACT

Aim. To study the relationship of karyotype, JAK2, CALR, and MPL driver mutations and ASXL1 mutation status with the progression and prediction of primary myelofibrosis (PMF).

Materials & Methods. The trial included 110 PMF patients (38 men and 72 women), median age was 59 years (range 18–82) with median follow-up after diagnosis of 2.6 years (range 0.1–23). The patients were examined for JAK2, CALR, MPL, and ASXL1 mutations. Restriction fragment length polymorphism technique was used for the analysis of V617F substitution in JAK2 and 515 codon mutation in MPL. CALR (exon 9) and ASXL1 (exon 12) mutation tests were performed using Sanger direct sequencing. In 48 (44 %) out of 110 patients bone marrow cell karyotype was determined. Clinical and hematological parameters and median overall survival (OS) of patients were analyzed with regard to detected genetic aberrations and combinations of them.

Results. JAK2, CALR, MPL mutations were detected in 55 (50 %), 28 (25.5 %), and 7 (6.4 %) out of 110 patients, respectively. Triple negative (TN) status was identified in 20 (18.2 %) out of 110 examined patients. ASXL1 mutations were detected in 22 (20 %) out of 110 patients. Out of 48 patients in 32 (66.7 %) normal karyotype, in 3 (6.3 %) favorable karyotype, in 4 (8.3 %) intermediate-prognosis karyotype, and in 9 (18.7 %) unfavorable karyotype were detected. The comparison of clinical and hematological parameters showed a number of significant differences. JAK2-positive patients had a higher hemoglobin level (median 129 g/L; = 0.021). TN was associated with a high IPSS risk (= 0.011), low hemoglobin level (median 101 g/L; = 0.006), continuing drop in platelet count (median 266 × 109/L; = 0.041), increased lymphocyte count (median 26.9 × 109/L; = 0.001). The detection of terminating mutations in ASXL1 correlated with palpable enlarged spleen (= 0.050), reduced platelet count (median 184 × 109/L; = 0.016), leukocyte count > 25 × 109/L (= 0.046), and blast count ≥ 1 % (< 0.001). Univariate regression analysis showed that terminating mutations in ASXL1 (hazard ratio [HR] 2.9; = 0.018), unfavorable karyotype (HR 8.2; < 0.001), and TN (ОР 8.1; < 0.001) had prognostic value for OS. ASXL1 mutation was associated with significantly worse OS in TN patients. Median OS of ASXL1-negative patients without high-risk chromosomal aberrations was significantly longer than in patients with high-risk karyotype and/or ASXL1 mutation.

Conclusion. Several genetic defects in tumor cells are associated with phenotypic manifestations of PMF. Based on the results of cytogenetic analysis and mutation determination of JAK2, CALR, MPL, and ASXL1, patients can be classified in different “genetic” risk groups when PMF is diagnosed.

Keywords: primary myelofibrosis, mutations, karyotype, prediction.

Received: April 8, 2019

Accepted: September 1, 2019

Read in PDF


REFERENCES

  1. Абдулкадыров К.М., Шуваев В.А., Мартынкевич И.С. Первичный миелофиброз: собственный опыт и новое в диагностике и лечении. Онкогематология. 2015;10(2):26–36. doi: 10.17650/1818-8346-2015-10-2-26-36.

    [Abdulkadyrov KM, Shuvaev VA, Martynkevich IS. Primary myelofibrosis: own experience and news from diagnostic and treatment. Oncohematology. 2015;10(2):26–36. doi: 10.17650/1818-8346-2015-10-2-26-36. (In Russ)]

  2. Абдулкадыров К.М., Шуваев В.А., Мартынкевич И.С. Миелопролиферативные новообразования. М.: Литтерра, 2016. 298 с.

    [Abdulkadyrov KM, Shuvaev VA, Martynkevich IS. Mieloproliferativnye novoobrazovaniya. (Myeloproliferative neoplasms.) Moscow: Litterra Publ.; 298 p. (In Russ)]

  3. Абдулкадыров К.М., Шуваев В.А., Мартынкевич И.С. Критерии диагностики и современные методы лечения первичного миелофиброза. Вестник гематологии. 2013;9(3):44–78.

    [Abdulkadyrov KM, Shuvaev VA, Martynkevich Diagnostic criteria and current methods of primary myelofibrosis treatment. Vestnik gematologii. 2013;9(3):44–78. (In Russ)]

  4. Tefferi A. Pathogenesis of myelofibrosis with myeloid metaplasia. J Clin Oncol. 2005;23(23):8520–30. doi: 10.1200/jco.2004.00.9316.

  5. Levine RL, Pardanani A, Tefferi A, et al. Role of JAK2 in the pathogenesis and therapy of myeloproliferative disorders. Nat Rev Cancer. 2007;7(9):673–83. doi: 10.1038/nrc2210.

  6. Milosevic Feenstra JD, Nivarthi H, Gisslinger H, et al. Whole-exome sequencing identifies novel MPL and JAK2 mutations in triple-negative myeloproliferative neoplasms. Blood. 2016;127(3):325–32. doi: 10.1182/blood-2015-07-661835.

  7. Tefferi A. Primary myelofibrosis: 2019 update on diagnosis, risk-stratification and management. Am J Hematol. 2018;93(12):1551–60. doi: 10.1002/ajh.25230.

  8. Tefferi A, Lasho TL, Finke CM, et al. Targeted deep sequencing in primary myelofibrosis. Blood Adv. 2016;1(2):105–11. doi: 10.1182/bloodadvances.2016000208.

  9. Hussein K, Van Dyke DL, Tefferi A. Conventional cytogenetics in myelofibrosis: literature review and discussion. Eur J Haematol. 2009;82(5):329–38. doi: 10.1111/j.1600-0609.2009.01224.x.

  10. Gangat N, Caramazza D, Vaidya R, et al. DIPSS Plus: A Refined Dynamic International Prognostic Scoring System for Primary Myelofibrosis That Incorporates Prognostic Information From Karyotype, Platelet Count, and Transfusion Status. J Clin Oncol. 2011;29(4):392–7. doi: 10.1200/jco.2010.32.2446.

  11. Guglielmelli P, Biamonte F, Score J, et al. EZH2 mutational status predicts poor survival in myelofibrosis. Blood. 2011;118(19):5227–34. doi: 10.1182/blood-2011-06-363424.

  12. Tefferi A, Lasho TL, Tischer A, et al. The prognostic advantage of calreticulin mutations in myelofibrosis might be confined to type 1 or type 1-like CALR Blood. 2014;124(15):2465–6. doi: 10.1182/blood-2014-07-588426.

  13. Tefferi A, Lasho TL, Finke C, et al. Type 1 vs type 2 calreticulin mutations in primary myelofibrosis: differences in phenotype and prognostic impact. Leukemia. 2014;28(7):1568–70. doi: 10.1038/leu.2014.83.

  14. Tefferi A, Guglielmelli P, Lasho TL, et al. CALR and ASXL1 mutations-based molecular prognostication in primary myelofibrosis: an international study of 570 patients. Leukemia. 2014;28(7):1494–500. doi: 10.1038/leu.2014.57.

  15. Argote JA, Dasanu CА. ASXL1 mutations in myeloid neoplasms: pathogenetic considerations, impact on clinical outcomes and survival. Curr Med Res Opin. 2016;34(5):757–63. doi: 10.1080/03007995.2016.1276896.

  16. Arber DA, Orazi A, Hasserjian R, et al. The 2016 revision of the World Health Organization classification of myeloid neoplasms and acute leukemia. Blood. 2016;127(20):2391–405. doi: 10.1182/blood-2016-03-643544.

  17. Guglielmelli P, Lasho TL, Rotunno G, et al. MIPSS70: Mutation-Enhanced International Prognostic Score System for Transplantation-Age Patients With Primary Myelofibrosis. J Clin Oncol. 2018;36(4):310–8. doi: 10.1200/jco.2017.76.4886.

  18. Tefferi A, Guglielmelli P, Lasho TL, et al. MIPSS70+ Version 2.0: Mutation and Karyotype Enhanced International Prognostic Scoring System for Primary Myelofibrosis. J Clin Oncol. 2018;36(17):1769–70. doi: 10.1200/jco.2018.78.9867.

  19. Tefferi A, Guglielmelli P, Nicolosi M, et al. GIPSS: genetically inspired prognostic scoring system for primary myelofibrosis. Leukemia. 2018;32(7):1631–42. doi: 10.1038/s41375-018-0107-z.

Cerebrovascular Disorders Associated with Ph-Negative Myeloproliferative Diseases

MM Tanashyan1, PI Kuznetsova1, AL Melikyan2, AA Raskurazhev1

1 Research Center of Neurology, 80 Volokolamskoe sh., Moscow, Russian Federation, 125367

2 National Medical Hematology Research Center, 4а Novyi Zykovskii pr-d, Moscow, Russian Federation, 125167

For correspondence: Polina Igorevna Kuznetsova, MD, PhD, 80 Volokolamskoe sh., Moscow, Russian Federation, 125367; Tel.: +7(495)490-24-05, +7(926)142-46-48; e-mail: angioneurology0@gmail.com

For citation: Tanashyan MM, Kuznetsova PI, Melikyan AL, Raskurazhev AA. Cerebrovascular Disorders Associated with Ph-Negative Myeloproliferative Diseases. Clinical oncohematology. 2019;12(4):398–405 (In Russ).

DOI: 10.21320/2500-2139-2019-12-4-398-405


ABSTRACT

Background. Cerebrovascular disorders continue to be among the most common and socially significant worldwide. Among multiple reasons for circulatory disturbances special importance is attached to hemorheology and hemostasis disorders occurring also in patients with Ph-negative myeloproliferative diseases (MPD).

Aim. To investigate typical characteristics of the course of cerebrovascular disorders in patients with Ph-negative MPD.

Materials & Methods. The trial included 169 adult patients with neurological diseases. Among them the main group consisted of 104 patients aged 34 to 55 years (median 48.5 years) with Ph-negative MPD diagnosed at the National Medical Hematology Research Center. The control group consisted of 65 patients aged 51 to 58 years (median 55.5 years) with cerebrovascular diseases without concomitant hematological pathology.

Results. The incidence of acute ischemic strokes was 26.2 % in polycythemia vera (PV), 20.5 % in essential thrombocythemia (ET), and 8.7 % in primary myelofibrosis (PMF).

Conclusion. An acute ischemic stroke with a concurrent thrombotic occlusion of one of the major head arteries is a criterion for ruling out Ph-negative MPD. To identify cerebral lesions in patients with Ph-negative MPD (PV, ET, PMF) MRI of the brain is recommended.

Keywords: cerebrovascular diseases, myeloproliferative diseases, thrombotic complications, hemorheology, hemostasis, ischemic stroke.

Received: February 13, 2019

Accepted: September 8, 2019

Read in PDF


REFERENCES

  1. Верещагин Н.В. Ангионеврология: гетерогенность ишемических нарушений мозгового кровообращения. В кн.: АМН СССР 60-я сессия. Л., 1990. С. 69–71.

    [Vereshchagin NV. Vascular neurology: heterogeneity of ischemic stroke. In: AMN SSSR 60-ya sessiya. (Academy of Medical Sciences of the USSR, the 60th session.) Leningrad; 1990. pp. 69–71. (In Russ)]

  2. Суслина З.А., Верещагин Н.В., Пирадов М.А. Подтипы ишемических нарушений мозгового кровообращения: диагностика и лечение. Consilium medicum. 2001;3(5):218–21.

    [Suslina ZA, Vereshchagin NV, Piradov MA. Subtypes of ischemic stroke: diagnosis and treatment. Consilium medicum. 2001;3(5):218–21. (In Russ)]

  3. Танашян М.М. Гемостаз, гемореология и атромбогенная активность сосудистой стенки в ангионеврологии. Анналы клинической и экспериментальной неврологии. 2007;1(2):29–33.

    [Tanashyan MM. Hemostasis, hemorheology and non-thrombogenic activity of vessel walls in vascular neurology. Annaly klinicheskoi i eksperimental’noi nevrologii. 2007;1(2):29–33. (In Russ)]

  4. Меликян А.Л., Туркина А.Г., Абдулкадыров К.М. и др. Клинические рекомендации по диагностике и терапии Ph-негативных миелопролиферативных заболеваний (истинная полицитемия, эссенциальная тромбоцитемия, первичный миелофиброз). Гематология и трансфузиология. 2014;59(4):31–56.

    [Melikyan AL, Turkina AG, Abdulkadyrov KM, et al. Clinical recommendations for the diagnosis and therapy of Ph-negative myeloproliferative diseases (polycythemia vera, essential thrombocythemia, primary myelofibrosis). Gematologiya i transfuziologiya. 2014;59(4):31–56. (In Russ)]

  5. Vardiman JW, Thiele J, Arber DA, et al. The 2008 revision of the World Health Organization (WHO) classification of myeloid neoplasms and acute leukemia: rationale and important changes. Blood. 2009;114(5):937–51. doi: 10.1182/blood-2009-03-209262.

  6. Arber DA, Orazi A, Hasserjian R, et al. The 2016 revision to the World Health Organization classification of myeloid neoplasms and acute leukemia. Blood. 2016;127(20):2391–405. doi: 10.1182/blood-2016-03-643544.

  7. Barbui T, Finazzi G, Carobbio A, et al. Development and validation of an International Prognostic Score of thrombosis in World Health Organization-essential thrombocythemia (IPSET-thrombosis). Blood. 2012;120(26):5128–33. doi: 10.1182/blood-2012-07-444067.

  8. Passamonti F, Rumi E, Pungolino E, et al. Life expectancy and prognostic factors for survival in patients with polycythemia vera and essential thrombocythemia. Am J Med. 2004;117(10):755–61. doi: 10.1016/j.amjmed.2004.06.032.

  9. Суборцева И.Н., Колошейнова Т.И., Пустовая Е.И. и др. Истинная полицитемия: обзор литературы и собственные данные. Клиническая онкогематология. 2015;8(4):397–412. doi: 10.21320/2500-2139-2015-8-4-397-412.

    [Subortseva IN, Kolosheinova TI, Pustovaya EI, et al. Polycythemia Vera: Literature Review and Own Data. Clinical oncohematology. 2015;8(4):397–412. doi: 10.21320/2500-2139-2015-8-4-397-412. (In Russ)]

  10. Танашян М.М., Кузнецова П.И., Суборцева И.Н., Меликян А.Л. Клинические особенности течения цереброваскулярной патологии при Ph-негативных миелопролиферативных заболеваниях. Клиническая фармакология и терапия. 2016;25(5):54–7.

    [Tanashyan MM, Kuznetsova PI, Subortseva IN, Melikyan AL. Clinical characteristics of the course of cerebrovascular pathology in Ph-negative myeloproliferative diseases. Klinicheskaya farmakologiya i terapiya. 2016;25(5):54–7. (In Russ)]

  11. Танашян М.М., Кузнецова П.И., Суборцева И.Н. и др. Хроническая и острая цереброваскулярная патология при Ph-негативных миелопролиферативных заболеваниях. Гематология и трансфузиология. 2016;61(3):146–50. doi: 10.18821/0234-5730-2016-61-3-146-150.

    [Tanashyan MM, Kuznetsova PI, Subortseva IN, et al. Chronic and acute cerebrovascular pathology in patients with Ph-negative myeloproliferative diseases. Gematologiya i transfuziologiya. 2016;61(3):146–50. doi: 10.18821/0234-5730-2016-61-3-146-150. (In Russ)]

  12. Duangnapasatit B, Rattarittamrong E, Rattanathammethee T, et al. Clinical Manifestations and Risk Factors for Complications of Philadelphia Chromosome-Negative Myeloproliferative Neoplasms. Asian Pac J Cancer Prev. 2015;16(12):5013–8. doi: 10.7314/apjcp.2015.16.12.5013.

  13. Tefferi A. Pathogenesis of myelofibrosis with myeloid metaplasia. J Clin Oncol. 2005;23(33):8520–30. doi: 10.1200/jco.2004.00.9316.

  14. Tanashyan MM, Kuznetsova PI, Shabalina AA, et al. Clinical characteristics of cerebrovascular pathology with patients suffering from Ph-negative myeloproliferative disease. Cerebrovasc Dis Extra. 2016;6(3):66–70. doi: 10.1159/000448597.

  15. Casini A, Fontana P, Lecompte T. Thrombotic complications of myeloproliferative neoplasms: risk assessment and risk-guided management. J Thromb Haemost. 2013;11(7):1215–27. doi: 10.1111/jth.12265.

  16. Wolanskyj AP, Schwager SM, McClure RF, et al. Essential thrombocythemia beyond the first decade: life expectancy, long-term complication rates, and prognostic factors. Mayo Clin Proc. 2006;81(2):159–66. doi: 10.4065/81.2.159.

  17. Carobbio A, Finazzi G, Guerini V, et al. Leukocytosis is a risk factor for thrombosis in essential thrombocythemia: interaction with treatment, standard risk factors, and Jak2 mutation status. Blood. 2007;109(6):2310–3. doi: 10.1182/blood-2006-09-046342.

  18. Меликян А.Л., Суборцева И.Н., Ковригина А.М. и др. Диагностика латентной истинной полицитемии (взгляд клинициста). Терапевтический архив. 2016;88(7):25–30. doi: 10.17116/terarkh201688725-30.

    [Melikian AL, Subortseva IN, Kovrigina AM, et al. Diagnosis of latent polycythemia vera: A clinician’s opinion. Terapevticheskii arkhiv. 2016;88(7):25–30. doi: 10.17116/terarkh201688725-30. (In Russ)]

  19. Vannucchi AM, Antonioli E, Guglielmelli P, et al. Clinical profile of homozygous JAK2 617V>F mutation in patients with polycythemia vera or essential thrombocythemia. Blood. 2007;110(3):840–6. doi: 10.1182/blood-2006-12-064287.

WT1 Gene Overexpression in Differential Diagnosis of Ph-negative Myeloproliferative Disorders

EG Lomaia1, NT Siordiya1, EG Lisina2, OM Senderova3, AA Silyutina1, AYu Zaritskey1

1 VA Almazov National Medical Research Center, 2 Akkuratova str., Saint Petersburg, Russian Federation, 197341

2 Municipal Clinical Hospital No. 52, 3 Pekhotnaya str., Moscow, Russian Federation, 123182

3 Irkutsk Regional Clinical Hospital, 100 Yubileinyi microdistrict, Irkutsk, Russian Federation, 664049

For correspondence: Nadiya Tamazovna Siordiya, 2 Akkuratova str., Saint Petersburg, Russian Federation, 197341; Tel.: +7(921)358-31-32; e-mail: siordian@list.ru

For citation: Lomaia EG, Siordiya NT, Lisina EG, et al. WT1 Gene Overexpression in Differential Diagnosis of Ph-Negative Myeloproliferative Disorders. Clinical oncohematology. 2019;12(3):297–302 (In Russ).

doi: 10.21320/2500-2139-2019-12-3-297-302


ABSTRACT

Aim. To assess the rate of WT1 gene overexpression and its clinical value in Ph-negative myeloproliferative disorders (MPD).

Materials & Methods. The trial included 72 patents with Ph-negative MPD. Among them there were patients with primary myelofibrosis (MF; n = 32), post-polycythemia vera MF (n = 7), polycythemia vera (PV; n = 17), and essential thrombocythemia (ET; n = 16) with median age of 57 years (range 19–78 years). Median (range) time from diagnosis to the date of evaluating WT1 expression in PV, ET, and MF was 9.4 (0–309), 14.4 (0–55), and 21.4 months (0–271 months), respectively. WT1 expression in terms of WT1 copies/104 ABL copies was measured by quantitative PCR.

Results. WT1 gene overexpression is revealed solely in patients with MF (in 34/39; 87 %). In PV/ET no WT1 gene overexpression was observed. Median WT1 expression in MF was 230/104 ABL copies (range 42.2–9,316.45/104 ABL copies). Sensitivity and specificity of WT1 gene overexpression in MF with respect to PV/ET were 87 % and 100 %, respectively. A distinct correlation was identified between WT1 gene expression level and spleen size, duration of the disease, blast cell count, and DIPSS risk group. WT1 gene expression level could be correlated neither with age and sex, nor with MF mutation status and leucocyte, thrombocyte, and haemoglobin levels.

Conclusion It appears that due to a high specificity and sensitivity of WT1 gene expression in MF it can be used as a marker for differential diagnosis of Ph-negative MPD. A correlation between WT1 gene expression and tumor mass in MF cannot be excluded. It is advisable to analyze the dynamics of WT1 expression level to predict the efficacy of current targeted therapy.

Keywords: WT1 gene, Ph-negative myeloproliferative disorders, myelofibrosis, polycythemia vera, essential thrombocythemia.

Received: December 27, 2018

Accepted: June 2, 2019

Read in PDF 


REFERENCES

  1. Han Y, San-Marina S, Liu J, et al. Transcriptional activation of c-myc proto-oncogene by WT1 protein. Oncogene. 2004;23(41):6933–41. doi: 10.1038/sj.onc.1207609.

  2. Hewitt SM, Hamada S, McDonnell TJ, et al. Regulation of the proto-oncogenes bcl-2 and c-myc by the Wilms’ tumor suppressor gene WT1. Cancer Res. 1995;55(22):5386–9.

  3. Jin DK, Kang SJ, Kim SJ, et al. Transcriptional regulation of PDGF-A and TGF-beta by +KTS WT1 deletion mutants and a mutant mimicking Denys-Drash syndrome. Ren Fail. 1999;21(6):685–94.

  4. Harrington MA, Konicek B, Song A, et al. Inhibition of colony-stimulating factor-1 promoter activity by the product of the Wilms’ tumor locus. J Biol Chem. 1993;268(28):21271–5.

  5. Hu Q, Gao F, Tian W, et al. Wt1 ablation and Igf2 upregulation in mice result in Wilms tumors with elevated ERK1/2 phosphorylation. J Clin Invest. 2011;121(1):174–83. doi: 10.1172/JCI43772

  6. Maurer U, Brieger J, Weidmann E, et al. The Wilms’ tumor gene is expressed in a subset of CD34+ progenitors and downregulated early in the course of differentiation in vitro. Exp Hematol. 1997;25(9):945–50.

  7. Baird PN, Simmons PJ. Expression of the Wilms’ tumor gene (WT1) in normal hemopoiesis. Exp Hematol. 1997;25(4):312–20.

  8. King-Underwood L, Renshaw J, Pritchard-Jones K. Mutations in the Wilms’ tumor gene WT1 in leukemias. Blood. 1996;87(6):2171–9.

  9. Ho PA, Zeng R, Alonzo TA, et al. Prevalence and prognostic implications of WT1 mutations in pediatric acute myeloid leukemia (AML): a report from the Children’s Oncology Group. Blood. 2010;116(5):702–10. doi: 10.1182/blood-2010-02-268953.

  10. Tamaki H, Ogawa H, Ohyashiki K, et al. The Wilms’ tumor gene WT1 is a good marker for diagnosis of disease progression of myelodysplastic syndromes. Leukemia. 1999;13(3):393–9. doi: 10.1038/sj.leu.2401341.

  11. Miwa H, Beran M, Saunders GF. Expression of the Wilms’ tumor gene (WT1) in human leukemias. 1992;6(5):405–9.

  12. Alberta JA, Springett GM, Rayburn H, et al. Role of the WT1 tumor suppressor in murine hematopoiesis. Blood. 2003;101(7):2570–4. doi: 10.1182/blood-2002-06-1656.

  13. Гиршова Л.Л., Будаева И.Г., Овсянникова Е.Г. и др. Прогностическое значение и корреляция динамики гиперэкспрессии гена WT1 и мутации гена NPM1 у пациентов с острым миелобластным лейкозом. Клиническая онкогематология. 2017;10(4):485–93. doi: 10.21320/2500-2139-2017-10-4-485-493.

    [Girshova LL, Budaeva IG, Ovsyannikova EG, et al. Prognostic Value and Correlation Between WT1 Overexpression and NPM1 Mutation in Patients with Acute Myeloblastic Leukemia. Clinical oncohematology. 2017;10(4):485–93. doi: 10.21320/2500-2139-2017-10-4-485-493. (In Russ)]

  14. Мамаев Н.Н., Гудожникова Я.В., Горбунова А.В. Гиперэкспрессия гена WT1при злокачественных опухолях системы крови: теоретические и клинические аспекты (обзор литературы). Клиническая онкогематология. 2016;9(3):257–64. doi: 10.21320/2500-2139-2016-9-3-257-264.

    [Mamaev NN, Gudozhnikova YaV, Gorbunova AV. WT1 Gene Overexpression in Oncohematological Disorders: Theoretical and Clinical Aspects (Literature Review). Clinical oncohematology. 2016;9(3):257–64. doi: 10.21320/2500-2139-2016-9-3-257-264. (In Russ)]

  15. Будаева И.Г., Гиршова Л.Л., Кузин С.О. и др. Прогностическое значение уровня гена WT1 у больных острыми миелоидными лейкозами с изолированной мутацией NPM1 и мутацией NPM1 c дополнительными молекулярными маркерами. Клиническая онкогематология. 2017;10(4):530–1.

    [Budaeva IG, Girshova LL, Kuzin SO, et al. Prognostic Value of WT1 Gene Level in Patients with Acute Myeloid Leukemia with Isolated NPM1 and NPM1 Mutation with Additional Molecular Markers. Clinical oncohematology. 2017;10(4):530–1. (In Russ)]

  16. Tamura H, Dan K, Yokose N, et al. Prognostic significance of WT1 mRNA and anti-WT1 antibody levels in peripheral blood in patients with myelodysplastic syndromes. Leuk Res. 2010;34(8):986–90. doi: 10.1016/j.leukres.2009.11.029.

  17. Gallo D, Nicoli P, Calabrese C, et al. The Wilms’ tumor (WT1) gene expression correlates with the International Prognostic Scoring System (IPSS) score in patients with myelofibrosis and it is a marker of response to therapy. Cancer Medicine. 2016;5(7):1650–3. doi: 10.1002/cam4.735.

  18. Siordiya N, Lisina E, Butylin P, et al. Incidence of Elevated Expression of wt1 in Primary Myelofibrosis (pmf) and Postpv-, Postet Myelofibrosis and Its Dynamics during Ruxolitinib Treatment. 2016;128:5498.

  19. Сиордия Н.Т., Булычева Е.Н., Холопова И.В. Частота встречаемости гиперэкспрессии WT1 у пациентов с миелоидными неоплазиями. Бюллетень Федерального Центра сердца, крови и эндокринологии им. В.А. Алмазова. 2012;6(17):116–20.

    [Siordiya NT, Bulycheva EN, Kholopova IV. WT1 overexpression rate in patents with myeloid neoplasm. Byulleten’ Federal’nogo Tsentra serdtsa, krovi i endokrinologii im. A. Almazova. 2012;6(17):116–20. (In Russ)]

  20. Cilloni D, Renneville A, Hermitte F, et al. Real-time quantitative polymerase chain reaction detection of minimal residual disease by standardized WT1 assay to enhance risk stratification in acute myeloid leukemia: A European Leukemia Net study. J Clin Oncol. 2009;27(31):5195–201. doi: 10.1200/jco.2009.22.4865.

  21. Vizmanos JL, Ormazabal C, Larrayoz MJ, et al. JAK2 V617F mutation in classic chronic myeloproliferative diseases: a report on a series of 349 patients. Leukemia. 2006;20(3):534–5. doi: 10.1038/sj.leu.2404086.

  22. Nangalia J, Massie CE, Baxter EJ, et al. Somatic CALR Mutations in Myeloproliferative Neoplasms with Nonmutated JAK2 N Engl J Med. 2013;369(25):2391–405. doi: 10.1056/NEJMoa1312542.

  23. Beer PA, Campbell PJ, Scott LM, et al. MPL mutations in myeloproliferative disorders: analysis of the PT-1 cohort. 2008;112(1):141–9. doi: 10.1182/blood-2008-01-131664.

  24. Меликян А.Л., Суборцева И.Н., Судариков А.Б. и др. Клинические особенности эссенциальной тромбоцитемии и первичного миелофиброза в зависимости от молекулярных характеристик заболевания. Терапевтический архив. 2017;89(7):4–9. doi: 10.17116/terarkh20178974-9.

    [Melikyan AL, Subortseva IN, Sudarikov AB, et al. Clinical features of essential thrombocythemia and primary myelofibrosis, depending on the molecular characteristics of disease. Terapevticheskii arkhiv. 2017;89(7):4–9. doi: 10.17116/terarkh20178974-9. (In Russ)]

  25. Жернякова А.А., Мартынкевич И.С., Шуваев В.А. и др. Молекулярно-генетические маркеры и особенности течения эссенциальной тромбоцитемии. Клиническая онкогематология. 2017;10(3):402–8. doi: 10.21320/2500-2139-2017-10-3-402-408.

    [Zhernyakova AA, Martynkevich IS, Shuvaev VA, et al. Molecular Genetic Markers and Clinical Characteristics of Essential Thrombocythemia. Clinical oncohematology. 2017;10(3):402–8. doi: 10.21320/2500-2139-2017-10-3-402-408. (In Russ)]

  26. Лисина Е.Г., Сиордия Н.Т., Бутылин П.А. и др. Клинико-лабораторные особенности эссенциального тромбоцитоза и первичного миелофиброза в зависимости от мутационного статуса генов JAK2 и CALR1. Онкогематология. 2017;12(3):8–16.

    [Lisina EG, Siordiya NT, Butylin PA, et al. Clinical and laboratory features of essential thrombocytosis and primary myelofibrosis depending on JAK2 and CALR1 mutation status. 2017;12(3):8–16. (In Russ)]

  27. Delic S, Rose D, Kern W, et al. Application of an NGS-based 28-gene panel in myeloproliferative neoplasms reveals distinct mutation patterns in essential thrombocythaemia, primary myelofibrosis and polycythaemia vera. Br J Haematol. 2016;175(3):419–26. doi: 10.1111/bjh.14269.

  28. Tefferi A. Novel mutations and their functional and clinical relevance in myeloproliferative neoplasms: JAK2, MPL, TET2, ASXL1, CBL, IDH and IKZF1. Leukemia. 2010;24(6):1128–38. doi: 10.1038/leu2010.69.

  29. Arber DA, Orazi A, Hasserjian R, et al. The 2016 revision to the World Health Organization classification of myeloid neoplasms and acute leukemia. Blood. 2016;127(20):2391–405. doi: 10.1182/blood-2016-03-643544.

  30. Меликян А.Л., Туркина А.Г., Ковригина А.М. и др. Клинические рекомендации по диагностике и терапии Ph-негативных миелопролиферативных заболеваний (истинная полицитемия, эссенциальная тромбоцитемия, первичный миелофиброз) (редакция 2016 г.). Гематология и трансфузиология. 2017;62(1, прил. 1):25–60.

    [Melikyan AL, Turkina AG, Kovrigina AM, et al. Clinical recommendations for diagnosis and therapy of Ph-negative myeloproliferative disorders (polycythemia vera, essential thrombocythemia, primary myelofibrosis) (edition 2016). Gematologiya i transfuziologiya. 2017;62(1, Suppl 1):25–60. (In Russ)]

  31. Bain BJ. Bone marrow biopsy morbidity: review of 2003. J Clin Pathol. 2005;58(4):406–8. doi: 10.1136/jcp.2004.022178.

  32. Arora B, Sirhan S, Hoyer JD, et al. Peripheral blood CD34 count in myelofibrosis with myeloid metaplasia: a prospective evaluation of prognostic value in 94 patients. Br J Haematol. 2005;128(1):42–8. doi: 10.1111/j.1365-2141.2004.05290.x.

  33. Barosi G, Viarengo G, Pecci A, et al. Diagnostic and clinical relevance of the number of circulating CD34+ cells in myelofibrosis with myeloidmetaplasia. Blood. 2001;98(12):3249–55. doi: 10.1182/blood.V98.12.3249.

  34. Xu M, Bruno E, Chao J, et al. Constitutive mobilization of CD34+ cells into the peripheral blood in idiopathic myelofibrosis may be due to the action of a number of proteases. Blood. 2005;105(11):4508–15. doi: 10.1182/blood-2004-08-3238.

  35. Забелина Т.С., Постриганева Т.И., Сайдали М.А. и др. Колониеобразующая способность клеток костного мозга и крови больных с различными формами лейкозов. Терапевтический архив. 1977;6:53–9.

    [Zabelina TS, Postriganeva TI, Saidali MA, et al. Bone marrow and blood cell colony-forming ability in patients with different leukemia types. Terapevticheskii arkhiv. 1977;6:53–9. (In Russ)]

  36. Harrison CN, Vannucchi AM, Kiladjian JJ, et al. Long-term findings from COMFORT-II, a phase 3 study of ruxolitinib vs best available therapy for myelofibrosis. Leukemia. 2016;30(8):1701–7. doi: 10.1038/leu.2016.148.

  37. Verstovsek S, Gupta V, Jason R, et al. A Pooled Overall Survival (OS) Analysis of 5-Year Data from the COMFORT-I and COMFORT-II Trials of Ruxolitinib for the Treatment of Myelofibrosis (MF). 2016;128(22):3110.

  38. Ионова Т.И., Анчукова Л.В., Виноградова О.Ю. и др. Качество жизни и спектр симптомов у больных миелофиброзом на фоне терапии: данные клинической практики. Гематология и трансфузиология. 2016;61(1):17–25.

    [Ionova TI, Anchukova LV, Vinogradova OYu, et al. Quality of life and symptoms in patients with myelofibrosis during the treatment: Data of clinical practice. Gematologiya i transfuziologiya. 2016;61(1):17–25. (In Russ)]

  39. Foltz L, Palumbo GA, Martino B, et al. Safety and Efficacy of Ruxolitinib for the Final Enrollment of JUMP: An Open-Label, Multicenter, Single-Arm, Expanded-Access Study in Patients with Myelofibrosis (n=2233). Blood. 2016;128(22):3107.

Prediction of FLAG ± Ida Regimen Efficacy in Patients with Relapsed/Refractory Acute Myeloid Leukemia

IG Budaeva, EG Ovsyannikova, EN Goryunova, OV Kulemina, DV Zaitsev, DV Motorin, RSh Badaev, DB Zammoeva, VV Ivanov, KV Bogdanov, OS Pisotskaya, YuV Mirolyubova, TS Nikulina, YuA Alekseeva, AYu Zaritskey, LL Girshova

VA Almazov National Medical Research Center, 2 Akkuratova str., Saint Petersburg, Russian Federation, 197341

For correspondence: Irina Garmaevna Budaeva, 2 Akkuratova str., Saint Petersburg, Russian Federation, 197341; Tel.: +7(931)351-07-06; e-mail: irina2005179@mail.ru

For citation: Budaeva IG, Ovsyannikova EG, Goryunova EN, et al. Prediction of FLAG ± Ida Regimen Efficacy in Patients with Relapsed/Refractory Acute Myeloid Leukemia. Clinical oncohematology. 2019;12(3):289-96 (In Russ).

doi: 10.21320/2500-2139-2019-12-3-289-296


ABSTRACT

Aim. To assess the efficacy of FLAG/FLAG-Ida regimen and to identify factors that influence remission, duration of disease-free survival (DFS) and overall survival (OS) of patients with relapsed/refractory acute myeloid leukemia (AML).

Materials & Methods. The trial included 54 patients (28 men and 26 women), median age was 37 years (range 18–70 years). 27 (50 %) out of 54 patients had refractory AML and 27 (50 %) patients had relapsed AML. FLAG and FLAG-Ida regimens were administered as induction therapy. 37 (68.5 %) patients received bone marrow transplantation. Molecular genetic and cytogenetic examinations were performed prior to therapy and on the 28th day after the start of treatment. WT1 gene expression was evaluated on the 14th–16th day of treatment.

Results. Complete remission (CR) was achieved in 42 (77.8 %) out of 54 patients. Refractoriness to therapy was observed in 9 (16.7 %) out of 54 patients, mortality was 5.5 % (3/54). Remission rate was higher in patients with relapsed AML compared with refractory AML: 85.2 % (23/27) and 70.4 % (19/27), respectively. On the 14th–16th day of treatment patients with blast cell count ≥ 10 % in bone marrow (BM) showed significantly lower CR rate (60 %) compared with the group of patients with < 10 % blast cells in BM (89.6 %; = 0.024) and shorter DFS (median 7.6 vs. 17.6 months, respectively; = 0.03). Median DFS in patients with WT1 expression reduction to < 1 log on the 14th–16th day was 5 vs. 18 months in patients without WT1 expression reduction (= 0.01). DFS varied in groups of patients with blast cell count < 10 % in BM on the 14th–16th day of treatment based on the level of WT1 expression reduction (= 0.04). MRD-negative patients (57.1 %) showed significantly longer DFS and OS compared with MRD-positive patients (median DFS was 17.6 vs. 5.2 months, respectively, = 0.02; median OS was 19 vs. 6.9 months, = 0.0002). Median DFS and OS were different only in ELN low- and high-risk groups (median not reached vs. 5.2 months, respectively, = 0.039; median not reached vs. 10.2 months, = 0.039).

Conclusion. FLAG and FLAG-Ida are effective and safe regimens in the treatment of relapsed/refractory AML. Achieving remission depends on neither the risk group nor the time of relapse occurrence. The blast cell count in BM on the 14th–16th day of FLAG/FLAG-Ida treatment is a prognostic factor determining achievement and duration of remission. WT1 expression level in the early post-induction period is a sensitive DFS marker. MRD status and molecular genetic risk (ELN) group affiliation are essential prognostic factors determining DFS and OS.

Keywords: acute myeloid leukemia, relapse, refractoriness, FLAG and FLAG-Ida regimens.

Received: November 2, 2018

Accepted: May 28, 2019

Read in PDF 


REFERENCES

  1. Papaemmanuil E, Gerstung M, Bullinger L. Genomic classification and prognosis in acute myeloid leukemia. N Engl J Med. 2016;374(23):2209–21. doi: 10.1056/NEJMoa1516192.

  2. Cheson BD, Bennett JM, Kopecky KJ, et al. Revised recommendations of the International Working Group for diagnosis, standardization of response criteria, treatment outcomes, and reporting standards for therapeutic trials in acute myeloid leukemia. J Clin Oncol. 2003;21(24):4642–9. doi: 10.1200/JCO.2003.04.036.

  3. Dohner H, Elihu H, Estey EH, et al. Diagnosis and management of acute myeloid leukemia in adults: recommendations from an international expert panel, on behalf of the European LeukemiaNet. Blood. 2010;115(3):453–74. doi: 10.1182/blood-2009-07-235358.

  4. Othus M, Appelbaum FR, Petersdorf SH, et al. Fate of patients with newly diagnosed acute myeloid leukemia who fail primary induction therapy. Biol Blood Marrow Transplant. 2015;21(3):559–64. doi: 10.1016/j.bbmt.2014.10.025

  5. Dohner H, Estey E, Grimwade D, et al. Diagnosis and management of AML in adults: 2017 ELN recommendations from an international expert panel. Blood. 2017;129(4):424–47. doi: 10.1182/blood-2016-08-733196.

  6. Elihu H, Estey E. Acute myeloid leukemia: 2016 Update on risk-stratification and management. Am J Hematol. 2016;91(8):824–46. doi: 10.1002/ajh.24439.

  7. Biggs JC, Horowitz MM, Gale RP, et al. Bone marrow transplants may cure patients with acute leukemia never achieving remission with chemotherapy. Blood. 1992;80(4):1090–3.

  8. Duval M, Klein JP, He W, et al. Hematopoietic stem-cell transplantation for acute leukemia in relapse or primary induction failure. J Clin Oncol. 2010;28(23):3730–8. doi: 10.1200/JCO.2010.28.8852.

  9. Sureda A. Indications for allo- and auto-SCT for hematological diseases, solid tumours and immune disorders: current practice in Europe. Bone Marrow Transplant. 2015;50(8):1037–56. doi: 10.1038/bmt.2015.6.

  10. Araki D, Othus M, Walter RB, et al. Effect of allogeneic hematopoietic cell transplantation in first complete remission on post-relapse complete remission rate and survival in acute myeloid leukemia. Haematologica. 2015;100(7):254–6. doi: 10.3324/haematol.2014.

  11. Delia M, Pastore D, Carluccio P, et al. FLAG-Ida regimen as bridge therapy to allotransplant in refractory/relapsed AML patients. Clin Lymph Myel Leuk. 2017;17(11):767–773. doi: 10.1016/j.clml.2017.06.002.

  12. Estey E, Kornblau S, Pierce S, et al. A stratification system for evaluating and selecting therapies in patients with relapsed or primary refractory acute myelogenous leukemia. Blood. 1996;88(2):756.

  13. Estey EH. Treatment of relapsed and refractory acute myelogenous leukemia. Leukemia. 2000;14(3):476–9. doi: 10.1038/sj.leu.2401568.

  14. Estey E, Plunkett W, Gandhi V, et al. Fludarabine and arabinosylcytosine therapy for refractory and relapsed acute myelogenous leukemia. Leuk Lymphoma. 1993;9(4–5):343–50. doi: 10.3109/10428199309148532.

  15. Estey E, Thall P, Andreeff M, et al. Use of granulocyte colony-stimulating factor before, during, and after fludarabine plus cytarabine induction therapy of newly diagnosed acute myelogenous leukemia or myelodysplastic syndromes; comparison with fludarabine plus cytarabine without granulocyte colony-stimulating factor. J Clin Oncol. 1994;12(4):671–8. doi: 10.1200/JCO.1994.12.4.671.

  16. Gandhi V, Plunkett W. Modulation of arabinosylnucleoside metabolism by arabinosylnucleotides in human leukemia cells. Cancer Res. 1988;48(2):329–34.

  17. Gandhi V, Estey E, Keating MJ, et al. Fludarabine potentiates metabolism of cytarabine in patients with acute myelogenous leukemia during therapy. J Clin Oncol. 1993;11(1):116–24. doi: 10.1200/JCO.1993.11.1.116.

  18. Anderlini P. Idarubicin cardiotoxicity: A retrospective study in acute myeloid leukemia and myelodysplasia. J Clin Oncol. 1995;13(11):2827–34. doi: 10.1200/JCO.1995.13.11.2827.

  19. Lee SR, Yang DH, Ahn JS, et al. The Clinical outcome of FLAG chemotherapy without idarubicin in patients with relapsed or refractory acute myeloid leukemia. J Korean Med Sci. 2009;24(3):498–503. doi: 10.3346/jkms.2009.24.3.498.

  20. Dohner H, Weisdorf DJ, Bloomfield CD. Acute Myeloid Leukemia. N Engl J Med. 2015;373(12):1136–52. doi: 10.1056/NEJMra1406184.

  21. Patel JP, Gonen M, Figueroa ME. Prognostic Relevance of Integrated Genetic Profiling in Acute Myeloid Leukemia. N Engl J Med. 2012;366(12):1079–89. doi: 10.1056/NEJMoa1112304.

  22. Wang LJ, Ding J, Zhu CY, et al. Clinic outcome of FLAG regimen treating patients with refractory and relapse acute myeloid leukemia. J Exper Hematol. 2016;24(1):19–24.

  23. Jun Xu, Ting-Ting Lv, Xiao-Fen Zhou, et al. Efficacy of common salvage chemotherapy regimens in patients with refractory or relapsed acute myeloid leukemia: A retrospective cohort study. Medicine. 2018;97(39): doi: 10.1097/MD.0000000000012102.

  24. Breems DA, Van Putten WL, Huijgens PC, et al. Prognostic index for adult patients with acute myeloid leukemia in first relapse. J Clin Oncol. 2005;23(9):1969–78. doi: 10.1200/jco.2005.06.027.

  25. Carella AM, Cascavilla N, Greco MM, et al. Treatment of poor risk acute myeloid leukemia with fludarabine, cytarabine and G-CSF (flag regimen): a single center study. Leuk Lymphoma. 2001;40(3–4):295–303. doi: 10.3109/10428190109057928.

  26. Ferrara F, Palmieri S, Pocali B, et al. De novo acute myeloid leukemia with multilineage dysplasia: treatment results and prognostic evaluation from a series of 44 patients treated with fludarabine, cytarabine and G-CSF (FLAG). Eur J Haematol. 2002;68(4):203–9. doi: 10.1034/j.1600-0609.2002.01651.x.

  27. Bao Y, Zhao J, Li Z-Z. Comparison of clinical remission and survival between CLAG and FLAG induction chemotherapy in patients with refractory or relapsed acute myeloid leukemia: a prospective cohort study. Clin Transl Oncol. 2018;20(7):870–80. doi: 10.1007/s12094-017-1798-8.

  28. Ossenkoppele GJ, Graveland WJ, Sonneveld P, et al. The value of fludarabine in addition to ARA-C and G-CSF in the treatment of patients with high-risk myelodysplastic syndromes and AML in elderly patients. Blood. 2004;103(8):2908–13. doi: 10.1182/blood-2003-07-2195.

  29. Jackson G, Taylor P, Smith GM, et al. A multicentre, open, non-comparative phase II study of a combination of fludarabine phosphate, cytarabine and granulocyte colony-stimulating factor in relapsed and refractory acute myeloid leukaemia and de novo refractory anaemia with excess of blasts in transformation. Br J Haematol. 2001;112(1):127–37. doi: 1046/j.1365-2141.2001.02551.x.

  30. Virchis A, Koh M, Rankin P, et al. Fludarabine, cytosine arabinoside, granulocyte-colony stimulating factor with or without idarubicin in the treatment of high risk acute leukaemia or myelodysplastic syndromes. Br J Haematol. 2004;124(1):26–32. doi: 10.1046/j.1365-2141.2003.04728.x.

  31. Farooq MU, Mushtaq F, Farooq A, et al. FLAG vs FLAG-IDA: outcomes in relapsed/refractory acute leukemias. Cancer Chemother Pharmacol. 2019;83(2):1–2. doi: 10.1007/s00280-019-03792-8.

  32. Heinemann V, Murray D, Walters R, et al. Mitoxantrone-induced DNA damage in leukemia cells is enhanced by treatment with high-dose arabinosylcytosine. Cancer Chemother Pharmacol. 1988;22(3):205–10. doi: 10.1007/BF00273412.

  33. Loughlin S, Gandhi V, Plunkett W, et al. The effect of 9-beta-D-arabinofuranosyl-2-fluoroadenine and 1-beta-D-arabinofuranosylcytosine on the cell cycle phase distribution, topoisomerase II level, mitoxantrone cytotoxicity, and DNA strand break production in K562 human leukemia cells. Cancer Chemother Pharmacol. 1996;38(3):261–8. doi: 10.1007/s002800050480.

  34. Gabert J, Beillard E, Velden VH, et al. Standardization and quality control studies of ‘real-time’ quantitative reverse transcriptase polymerase chain reaction of fusion gene transcripts for residual disease detection in leukemia – a Europe Against Cancer program. Leukemia. 2003;17(12):2318–57. doi: 10.1038/sj.leu.2403135.

  35. Willasch AM, Gruhn B, Coliva T, et al. Combined usage of Wilms’ tumor gene quantitative analysis and multiparameter flow cytometry for minimal residual disease monitoring of acute myeloid leukemia patients after allogeneic hematopoietic stem cells transplantation. Exp Ther Med. 2018;15(2):1403–9. doi: 10.3892/etm.2017.5547.

  36. Богданов К.В., Моторин Д.В., Никулина Т.С. и др. Мониторинг донорского химеризма и минимальной остаточной болезни у онкогематологических больных после аллогенной трансплантации гемопоэтических стволовых клеток. Биомедицинская химия. 2017;63(6):570–81. doi: 10.18097/PBMC

    [Bogdanov KV, Motorin DV, Nikulina TS, et al. Donor chimerism and minimal residual disease monitoring in leukemia patients after allo-HSCT. Biomeditsinskaya khimiya. 2017;63(6):570–81. doi: 10.18097/PBMC20176306570. (In Russ)]

  37. Pastore D, Specchia G, Carluccio P, et al. FLAG-IDA in the treatment of refractory/relapsed acute myeloid leukemia: single-center experience. Ann Hematol. 2003;82(4):231–5. doi: 10.1007/s00277-003-0624-2.

  38. Montillo M, Mirto S, Petti MC, et al. Fludarabine, cytarabine, and G-CSF (FLAG) for the treatment of poor risk acute myeloid leukemia. Am J Hematol. 1998;58(2):105–9. doi: 1002/(sici)1096-8652(199806)58:2<105::aid-ajh3>3.0.co;2-w.

  39. Nokes TJ, Johnson S, Harvey D, et al. FLAG is a useful regimen for poor prognosis adult myeloid leukaemias and myelodysplastic syndromes. Leuk Lymphoma. 1997;27(1–2):93–101. doi: 10.3109/10428199709068275.

Prognostic Value of Genetic Mutations in Patients with Acute Myeloid Leukemias: Results of a Cooperative Study of Hematology Clinics of Saint Petersburg (Russia) and Charite Clinic (Germany)

EV Motyko1, OV Blau2, LB Polushkina1, LS Martynenko1, MP Bakai1, NYu Tsybakova1, YuS Ruzhenkova1, EV Kleina1, NB Pavlenko1, AM Radzhabova1, EV Karyagina3, OS Uspenskaya4, SV Voloshin1, AV Chechetkin1, IS Martynkevich1

1 Russian Research Institute of Hematology and Transfusiology, 16 2-ya Sovetskaya str., Saint Petersburg, Russian Federation, 191024

2 Charite Clinic, Berlin Medical University, 30 Hindenburgdamm, Berlin, Germany, 12200

3 Municipal Hospital No. 15, 4 Avangardnaya str., Saint Petersburg, Russian Federation, 198205

4 Leningrad Regional Clinical Hospital, 45–49 Lunacharskogo pr-t, Saint Petersburg, Russian Federation, 194291

For correspondence: Ekaterina Vadimovna Motyko, PhD in Biology, 16 2-ya Sovetskaya str., Saint Petersburg, Russian Federation, 191024; Tel.: +7(812)925-05-62; e-mail: genetics.spb@mail.ru

For citation: Motyko EV, Blau OV, Polushkina LB, et al. Prognostic Value of Genetic Mutations in Patients with Acute Myeloid Leukemias: Results of a Cooperative Study of Hematology Clinics of Saint Petersburg (Russia) and Charite Clinic (Germany). Clinical oncohematology. 2019;12(2):211–9.

DOI: 10.21320/2500-2139-2019-12-2-211-219


ABSTRACT

Aim. To analyze the effect on prognosis of mutations that are typical of acute myeloid leukemia (AML) patients.

Materials & Methods. The study included 620 AML patients surveyed at Hematology Clinics of Saint Petersburg (Russia) and Charite Clinic (Berlin, Germany). G-banding of chromosomes was employed for cytogenetic testing. Aberration screening in DNMT3A, IDH1/2 genes was based on real-time polymerase chain reaction (PCR) with subsequent analysis of melting and sequencing profiles. Mutations in FLT3, NPM1 genes were revealed by PCR.

Results. Mutations were identified in 343 (55.3 %) out of 620 patients. Significantly more often mutations were discovered in patients with normal karyotype (NK) (= 0.001). FLT3-ITD mutation was associated with reduced medians of overall survival (OS) and disease-free (DFS) survival: 11.3 vs. 15.8 months with FLT3-ITD– (= 0.005) and 10.0 vs. 13.3 months with FLT3-ITD+ (= 0.009), respectively. The relation of FLT3-ITD allele burden to OS duration was also assessed. In the ITDlow/ITD– group the OS median was considerably longer than in the ITDhigh group (= 0.028). In the group of patients with 1 mutation in NPM1 gene OS and DFS were much better in comparison with other patients (medians of 27.4 and 13.9 months, respectively, = 0.040; 19.3 and 12.0 months, = 0.049). Negative impact of mutations in DNMT3A gene was noticed while assessing OS median: 12 (DNMT3A+) and 15 months (DNMT3A–), respectively (= 0.112). Mutations in IDH1 gene correlated with a better OS than in the group without mutations (= 0.092). The rs11554137 polymorphism in IDH1 gene was associated with worse OS in the group of patients with NK (= 0.186). In 144 patients various mutation combinations (from 2 to 5) were identified. It was demonstrated that mutations in FLT3 (FLT3-ITD), NPM1, DNMT3A, and IDH2 were identified significantly more often in combinations with other mutations (= 0.001): NPM1+/FLT3-ITD+ (20.8 %), NPM1+/FLT3-ITD+/DNMT3A+ (8.3 %), and FLT3-ITD+/DNMT3A+ (8.3 %). Patients with 1 mutation had a noticeably longer OS median compared with patients with 2 mutations (18.1 and 12.2 months; = 0.003). In patients with NPM1+ according to their OS the most unfavorable additional mutation was FLT3-ITD (median 27.4 vs. 9.2 months; = 0.019) and the combination of NPM1+/FLT3-ITD+/DNMT3A+ (median 27.4 vs. 14.6 months; = 0.141). OS of patients with DNMT3A+ showed a downward trend if FLT3-ITD additional mutation was identified (17.3 vs. 7.1 months; = 0.074).

Conclusion. Mutations in FLT3, DNMT3A, IDH1/2, NPM1 genes frequently occur in AML intermediate-risk patients, i.e. they determine the intermediate prognosis group in AML. The studied mutations considerably impact prognosis. It is important to take into consideration mutation type, its allele burden, and the presence of additional mutations. A patient with 2 mutations has a considerably worse OS compared with a patient with 1 mutation. The studied group of patients with the combination of NPM1+/FLT3-ITD+, NPM1+/FLT3-ITD+/DNMT3A+, DNMT3A+/FLT3-ITD+ mutations has the poorest prognosis. Comprehensive analysis of genetic damages in AML patients allows to most accurately predict the course and prognosis of the disease and to plan targeted therapy.

Keywords: acute myeloid leukemias, mutations in FLT3, NPM1, DNMT3A, IDH1/2 genes, karyotype, prognosis.

Received: July 13, 2018

Accepted: January 16, 2019

Read in PDF 


REFERENCES

  1. Schlenk RF, Dohner H. Genomic applications in the clinic: use in treatment paradigm of acute myeloid leukemia. Hematol Am Soc Hematol Educ Program. 2013;2013(1):324–30. doi: 10.1182/asheducation-2013.1.324.

  2. Sanders MA, Valk PJ. The evolving molecular genetic landscape in acute myeloid leukaemia. Curr Opin Hematol. 2013;20(2):79–85. doi: 10.1097/MOH.0b013e32835d821c.

  3. Preisler H, Davis RB, Kirshner J, et al. Comparison of three remission induction regimens and two postinduction strategies for the treatment of acute nonlymphocytic leukemia: a cancer and leukemic group B study. Blood. 1987;69(5):1441–9.

  4. Wiernik PH, Banks PLC, Case DC, et al. Cytarabine plus idarubicin or daunorubicin as induction and consolidation therapy for previously untreated adult patients with acute myeloid leukemia. 1992;79(2):313–9.

  5. Алгоритмы диагностики и протоколы лечения заболеваний системы крови. Под ред. В.Г. Савченко. М.: Практика, 2018. Т. 1. 1008 с.

    [Savchenko VG, ed. Algoritmy diagnostiki i protokoly lecheniya zabolevanii sistemy krovi. (Diagnostic algorithms and treatment protocols for blood system diseases.) Moscow: Praktika Publ.; 2018. Vol. 1. 1008 p. (In Russ)]

  6. Bennett JM, Catovsky D, Daniel MT, et al. Proposals for the classification of the acute leukaemias. French-American-British (FAB) co-operative group. Br J Haematol. 1976;33(4):451–8. doi: 10.1111/j.1365-2141.1976.tb03563.x.

  7. Heim S, Mitelman F. Cancer Cytogenetics: chromosomal and molecular genetic aberrations of tumor cells. 4th ed. Wiley-Blackwell: 2015. рр. 632. doi: 10.1002/9781118795569.

  8. Jordan CT. Unique molecular and cellular features of acute myelogenous leukemia stem cells. 2002;16(4):559–62. doi: 10.1038/sj.leu.2402446.

  9. Ding L, Ley TJ, Larson DE, et al. Clonal evolution in relapsed acute myeloid leukaemia revealed by whole-genome sequencing. Nature. 2012;481(7382):506– doi: 10.1038/nature10738.

  10. Gerlinger M, Rowan AJ, Horswell S, et al. Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. N Engl J Med. 2012;366(10):883–92. doi: 10.1056/NEJMoa1113205.

  11. Campbell PJ, Pleasance ED, Stephens PJ, et al. Subclonal phylogenetic structures in cancer revealed by ultra-deep sequencing. Proc Natl Acad Sci USA. 2008;105(35):13081–6. doi: 10.1073/pnas.0801523105.

  12. Kottaridis PD, Gale RE, Frew ME, et al. The presence of a FLT3 internal tandem duplication in patients with acute myeloid leukemia (AML) adds important prognostic information to cytogenetic risk group and response to the first cycle of chemotherapy: analysis of 854 patients from the United Kingdom Medical Research Council AML 10 and 12 trials. Blood. 2001;98(6):1752– doi: 10.1182/blood.v98.6.1752.

  13. Santos FP, Jones D, Qiao W, et al. Prognostic value of FLT3 mutations among different cytogenetic subgroups in acute myeloid leukemia. Cancer. 2011;117(10):2145–55. doi: 10.1002/cncr.25670.

  14. Sallman DA, Lancet JE. What are the most promising new agents in acute myeloid leukemia? Curr Opin Hematol. 2017;24(2):99–107. doi: 10.1097/MOH.0000000000000319.

  15. Thiede C, Koch S, Creutzig E, et al. Prevalence and prognostic impact of NPM1 mutations in 1485 adult patients with acute myeloid leukemia (AML). 2006;107(10):4011–20. doi: 10.1182/blood-2005-08-3167.

  16. Dohner K, Schlenk RF, Habdank M, et al. Mutant nucleophosmin (NPM1) predicts favorable prognosis in younger adults with acute myeloid leukemia and normal cytogenetics: interaction with other gene mutations. Blood. 2005;106(12):3740–6. doi: 10.1182/blood-2005-05-2164.

  17. Тилова Л.Р., Савинкова А.В., Жидкова Е.М. и др. Молекулярно-генетические нарушения в патогенезе опухолей системы крови и соответствующие им изменения сигнальных систем клетки. Клиническая онкогематология. 2017;10(2):235– doi: 10.21320/2500-2139-2017-10-2-235-249.

    [Tilova LR, Savinkova AV, Zhidkova EM, et al. Molecular Genetic Abnormalities in the Pathogenesis of Hematologic Malignancies and Corresponding Changes in Cell Signaling Systems. Clinical oncohematology. 2017;10(2):235–49. doi: 10.21320/2500-2139-2017-10-2-235-249. (In Russ)]

  18. Emadi A, Faramand R, Carter-Cooper B, et al. Presence of isocitrate dehydrogenase mutations may predict acute myeloid leukemia. Am J Hematol. 2015;90(5):E77–9. doi: 10.1002/ajh.23965.

  19. Patel JP, Gonen M, Figueroa ME, et al. Prognostic relevance of integrated genetic profiling in acute myeloid leukemia. N Engl J Med. 2012;366(12):1079–89. doi: 10.1056/NEJMoa1112304.

  20. Renneville A, Boissel N, Nibourel O, et al. Prognostic significance of DNA methyltransferase 3A mutations in cytogenetically normal acute myeloid leukemia: a study by the Acute Leukemia French Association. Leukemia. 2012;26(6):1247–54. doi: 10.1038/leu.2011.382.

  21. Marcucci G, Maharry K, Wu Y-Z, et al. IDH1 and IDH2 gene mutations identify novel molecular subsets within de novo cytogenetically normal acute myeloid leukemia: a Cancer and Leukemia Group B study. J Clin Oncol. 2010;28(14):2348–55. doi: 10.1200/JCO.2009.27.3730.

  22. Paschka P, Schlenk RF, Gaidzik VI, et al. IDH1 and IDH2 mutations are frequent genetic alterations in acute myeloid leukemia and confer adverse prognosis in cytogenetically normal acute myeloid leukemia with NPM1 mutation without FLT3 internal tandem duplication. J Clin Oncol. 2010;28(22):3636–43. doi: 10.1200/JCO.2010.28.3762.

  23. Abbas S, Lugthart S, Kavelaars FG, et al. Acquired mutations in the genes encoding IDH1 and IDH2 both are recurrent aberrations in acute myeloid leukemia: prevalence and prognostic value. Blood. 2010;116(12):2122–6. doi: 10.1182/blood-2009-11-250878.

  24. Thol F, Damm F, Ludeking A, et al. Incidence and prognostic influence of DNMT3A mutations in acute myeloid leukemia. J Clin Oncol. 2011;29(21):2889– doi: 10.1200/JCO.2011.35.4894.

  25. Ley TJ, Miller C, Ding L, Raphael BJ, et al. Genomic and epigenomic landscapes of adult de novo acute myeloid leukemia. N Engl J Med. 2013;368(22):2059– doi: 10.1056/NEJMoa1301689.

  26. Kihara R, Nagata Y, Kiyoi H, et al. Comprehensive analysis of genetic alterations and their prognostic impacts in adult acute myeloid leukemia patients. Leukemia. 2014;28(8):1586– doi: 10.1038/leu.2014.55.

  27. Ravandi F, Kantarjian H, Faderl S, et al. Outcome of patients with FLT3-mutated acute myeloid leukemia in first relapse. Leuk Res. 2010;34(6):752– doi: 10.1016/j.leukres.2009.10.001.

  28. Frohling S, Schlenk RF, Breitruck J, et al. Prognostic significance of activating FLT3 mutations in younger adults (16 to 60 years) with acute myeloid leukemia and normal cytogenetics: A study of the AML study group Ulm. Blood. 2002;100(13):4372– doi: 10.1182/blood-2002-05-1440.

  29. Schlenk RF, Kayser S, Bullinger L, et al. Differential impact of allelic ratio and insertion site in FLT3-ITD–positive AML with respect to allogeneic transplantation. Blood. 2014;124(23):3441– doi: 10.1182/blood-2014-05-578070.

  30. Kim Y, Lee GD, Park J, et al. Quantitative fragment analysis of FLT3-ITD efficiently identifying poor prognostic group with high mutant allele burden or long ITD length. Blood Cancer J. 2015;5(8):e336. doi: 10.1038/bcj.2015.61.

  31. Linch DC, Hills RK, Burnett AK, et al. Impact of FLT3ITD mutant allele level on relapse risk in intermediate-risk acute myeloid leukemia. Blood. 2014;124(2):273– doi: 10.1182/blood-2014-02-554667.

  32. Brunet S, Labopin M, Esteve J, et al. Impact of FLT3 internal tandem duplication on the outcome of related and unrelated hematopoietic transplantation for adult acute myeloid leukemia in first remission: a retrospective analysis. J Clin Oncol. 2012;30(7):735– doi: 10.1200/JCO.2011.36.9868.

  33. DeZern AE, Sung A, Kim S, et al. Role of allogeneic transplantation for FLT3/ITD acute myeloid leukemia: outcomes from 133 consecutive newly diagnosed patients from a single institution. Biol Blood Marrow Transplant. 2011;17(9):1404– doi: 10.1016/j.bbmt.2011.02.003.

  34. Islam M, Mohamed Z, Assenov Y. Differential analysis of genetic, epigenetic, and cytogenetic abnormalities in AML. Int J Genom. 2017;2017:2913648. doi: 10.1155/2017/2913648.

  35. Papaemmanuil E, Gerstung M, Bullinger L, et al. Genomic classification and prognosis in acute myeloid leukemia. N Engl J Med. 2016;375(9):900– doi: 10.1056/NEJMc1608739.

  36. Dohner H, Estey E, Amadori S, et al. Diagnosis and Management of Acute Myeloid Leukemia in Adults: Recommendations from an International Expert Panel, on Behalf of the European LeukemiaNet. Blood. 2010;115(3):453– doi: 10.1182/blood-2009-07-235358.

  37. Gale RE, Green C, Allen C, et al. The impact of FLT3 internal tandem duplication mutant level, number, size, and interaction with NPM1 mutations in a large cohort of young adult patients with acute myeloid leukemia. Blood. 2008;111(5):2776– doi: 10.1182/blood-2007-08-109090.

  38. Pratcorona M, Brunet S, Nomdedeu J, et al. Favorable outcome of patients with acute myeloid leukemia harboring a low-allelic burden FLT3-ITD mutation and concomitant NPM1 mutation: Relevance to post-remission therapy. Blood. 2013;121(14):2734– doi: 10.1182/blood-2012-06-431122.

  39. Stone RM, Mandrekar S, Sanford BL, et al. The Multi-Kinase Inhibitor Midostaurin (M) Prolongs Survival Compared with Placebo (P) in Combination with Daunorubicin (D)/Cytarabine (C) Induction (ind), High-Dose C Consolidation (consol), and As Maintenance (maint) Therapy in Newly Diagnosed Acute Myeloid Leukemia (AML) Patients (pts) Age 18–60 with FLT3 Mutations (muts): An International Prospective Randomized (rand) P-Controlled Double- Blind Trial (CALGB 10603/RATIFY [Alliance]). Blood. 2015;126(23): 6, abstract.

  40. Ibrahem L, Mahfouz R, Elhelw L, et al. Prognostic significance of DNMT3A mutations in patients with acute myeloid leukemia. Blood Cells Mol Dis. 2015;54(1):84– doi: 10.1016/j.bcmd.2014.07.015.

  41. Ley T, Ding L, Walter M, et al. DNMT3A mutations in acute myeloid leukemia. N Engl J Med. 2010;363(25):2424– doi: 10.1056/NEJMoa1005143.

  42. Willander K, Falk I, Chaireti R, et al. Mutations in the isocitrate dehydrogenase 2 gene and IDH1 SNP 105C>T have a prognostic value in acute myeloid leukemia. Biomark Res. 2014;2(1):18. doi: 10.1186/2050-7771-2-18.

  43. Xu Q, Li Y, Lv N, et al. Correlation between isocitrate dehydrogenase gene aberrations and prognosis of patients with acute myeloid leukemia: a systematic review and meta-analysis. Clin Cancer Res. 2017;23(15):4511– doi: 10.1158/1078-0432.CCR-16-2628.

  44. Wagner K, Damm F, Gohring G, et al. Impact of IDH1 R132 mutations and an IDH1 single nucleotide polymorphism in cytogenetically normal acute myeloid leukemia: SNP rs11554137 is an adverse prognostic factor. J Clin Oncol. 2010;28(14):2356– doi: 10.1200/JCO.2009.27.6899.

  45. Stein EM, Tallman MS. Emerging therapeutic drugs for AML. Blood. 2016;127(1):71– doi: 10.1182/blood-2015-07-604538.

  46. Ploen GG, Nederby L, Guldberg P, et al. Persistence of DNMT3A mutations at long-term remission in adult patients with AML. Br J Haematol. 2014;167(4):478– doi: 10.1111/bjh.13062.

  47. Gaidzik V, Weber D, Paschka P, et al. Monitoring of minimal residual disease (MRD) of DNMT3A mutations (DNMT3Amut) in acute myeloid leukemia (AML): a study of the AML Study Group (AMLSG). Blood. 2015;126(23):226, abstract.

Management of Chronic Myeloid Leukemia Patients During Pregnancy (Analysis of Literature and Practical Recommendations)

EYu Chelysheva1, AG Turkina1, ES Polushkina2, MA Vinogradova2, RG Shmakov2

1 National Medical Hematology Research Center, 4a Novyi Zykovskii pr-d, Moscow, Russian Federation, 125167

2 VI Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, 4 Akademika Oparina str., Moscow, Russian Federation, 117997

For correspondence: Ekaterina Yur’evna Chelysheva, MD, PhD, 4a Novyi Zykovskii pr-d, Moscow, Russian Federation, 125167; Tel.: +7(495)612-48-60; e-mail: denve@bk.ru

For citation: Chelysheva EYu, Turkina AG, Polushkina ES, et al. Management of Chronic Myeloid Leukemia Patients During Pregnancy (Analysis of Literature and Clinical Experience). Clinical oncohematology. 2019;12(2):202–10.

DOI: 10.21320/2500-2139-2019-12-2-202-210


ABSTRACT

Background. The tyrosine kinase inhibitors (TKI) era is marked by a long-term favorable prognosis of chronic myeloid leukemia (CML). In this context CML patients of reproductive age are faced with major issues of family planning with due regard to the risk of TKI treatment interruption during pregnancy. Additionally, TKI impact is another potential risk to the fetus.

Aim. To develop differentiated approach to CML treatment during pregnancy.

Materials & Methods. Analysis includes literature data and clinical experience based on 166 pregnancies of 120 CML patients from CML Pregnancy Registry.

Results. Pregnancy planning is recommended after achieving stable and deep molecular response (with BCR-ABL > 0.01 %, IS) within the period of at least 2 years. At conception TKI therapy does not have to be interrupted. However, early pregnancy detection and TKI treatment interruption after pregnancy confirmation are of vital importance due to teratogenic risks. Furthermore, no TKI may be administered during organogenetic period, i.e. up to the 15th week of gestation. In the absence or loss of complete hematologic response and growth of BCR-ABL > 1 % after the 15th week of gestation imatinib or nilotinib administration is justified in the interest of pregnant patients taking into account limited transfer of these drugs through placenta. In the absence of complete hematologic response before the 15th week of gestation interferon-α can be administered. With BCR-ABL < 1 % patients can be either followed-up without therapy or they can receive interferon-α throughout pregnancy. Dasatinib, bosutinib, and other TKI are contraindicated at any stage of pregnancy. There are no special recommendations for childbirth, delivery is to be adapted to obstetric conditions. Breast feeding is not recommended because of the lack of practical evidence for its safety.

Conclusion. A regular molecular monitoring of BCR-ABL and hematologic status is indispensable, health condition of fetus should be continuously monitored as well. CML patient management should be conducted by cooperating hematologists and gynecologists.

Keywords: chronic myeloid leukemia, pregnancy, tyrosine kinase inhibitors, imatinib, nilotinib, dasatinib, bosutinib.

Received: January 9, 2019

Accepted: March 20, 2019

Read in PDF 


REFERENCES

  1. Hochhaus A, Larson RA, Guilhot F, et al. Long-term outcomes of imatinib treatment for chronic myeloid leukemia. N Engl J Med. 2017;376(10):917–27. doi: 10.1056/NEJMoa1609324.

  2. Shukhov O, Chelysheva E, Gusarova G, et al. Imatinib treatment in chronic myeloid leukemia patients in early and late chronic phase: current incidence of cytogenetic remission and a very long-term an intention-to-treat analysis. Haematologica. 2015;100(Suppl 1):437.

  3. Куликов С.М., Виноградова О.Ю., Челышева Е.Ю. и др. Заболеваемость хроническим миелолейкозом в 6 регионах России по данным популяционного исследования 2009–2012 гг. Терапевтический архив. 2014;86(7):24–30.

    [Kulikov SM, Vinogradova OYu, Chelysheva EYu, et al. Incidence of chronic myeloid leukemia in 6 regions of Russia according to the data of the 2009–2012 population-based study. Terapevticheskii arkhiv. 2014;86(7):24–30. (In Russ)]

  4. Hoffmann VS, Baccarani M, Hasford J, et al. The EUTOS population-based registry: incidence and clinical characteristics of 2904 CML patients in 20 European countries. Leukemia. 2015;29(6):1336–43. doi: 10.1038/leu.2015.73.

  5. Carlier P, Bernard N, Lagarce L, et al. Pregnancy outcome among partners of male patients receiving imatinib, dasatinib or nilotinib in chronic myeloid leukemia: reports collected by the French network pharmacovigilance centers. Arch Gynecol Obstet. 2017;295(2):269–71. doi: 10.1007/s00404-016-4262-z.

  6. Abruzzese E, Trawinska MM, de Fabritiis P, et al. Tyrosine kinase inhibitors and pregnancy. Mediterr J Hematol Infect Dis. 2014;6(1):2014028. doi: 10.4084/MJHID.2014.028.

  7. Cortes JE, Gambacorti-Passerini C, Deininger MW, et al. Pregnancy outcomes in patients treated with bosutinib. Blood. 2018;132:1729, abstract.

  8. Palani R, Milojkovic D, Apperley JF. Managing pregnancy in chronic myeloid leukemia. Ann Hematol. 2015;94(Suppl 2):S167–76. doi: 10.1007/s00277-015-2317-z.

  9. Bhandari A, Rolen K, Shah BK. Management of chronic myelogenous leukemia in pregnancy. Anticancer Res. 2015;35(1):1–11.

  10. Abruzzese E, Trawinska MM, de Fabritiis P, et al. Management of pregnant chronic myeloid leukemia patients. Expert Rev Hematol. 2016;9(8):781–91. doi: 10.1080/17474086.2016.1205479.

  11. Челышева Е.Ю., Туркина А.Г. Протокол лечения хронического миелолейкоза во время беременности. В кн.: Алгоритмы диагностики и протоколы лечения заболеваний системы крови. Под ред. В.Г. Савченко. М.: Практика, 2018. Т. 2. С. 927–49.

    [Chelysheva EYu, Turkina AG. Protocol of chronic myeloid leukemia treatment during pregnancy. In: Savchenko VG, ed. Algoritmy diagnostiki i protokoly lecheniya zabolevanii sistemy krovi. (Diagnostic algorithms and treatment protocols for blood system diseases.) Moscow: Praktika Publ.; 2018. Vol. 2. pp. 927–49. (In Russ)]

  12. Челышева Е.Ю., Туркина А.Г., Чабаева Ю.А. и др. Регистр случаев беременности при хроническом миелолейкозе: клинико-демографическая характеристика пациентов. Гематология и трансфузиология. 2016;61(1-S1):79.

    [Chelysheva EYu, Turkina AG, Chabaeva YuA, et al. Registry of pregnancy cases in chronic myeloid leukemia: clinical and demographic characteristics of patients. Gematologiya i transfuziologiya, 2016;61(1-S1):79. (In Russ)]

  13. Baccarani M, Deininger MW, Rosti G, et al. European LeukemiaNet recommendations for the management of chronic myeloid leukemia: 2013. Blood. 2013;122(6):872–84. doi: 10.1182/blood-2013-05-501569.

  14. Hughes TP, Ross DM. Moving treatment-free remission into mainstream clinical practice in CML. 2016;128(1):17–23. doi: 10.1182/blood-2016-01-694265.

  15. Hochhaus A, Saussele S, Rosti G, et al. Chronic myeloid leukaemia: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2018;29(Suppl 4):iv261. doi: 10.1093/annonc/mdy159.

  16. National Comprehensive Cancer Network. Chronic myeloid leukemia (Version 4.2018). Available from: https://www.nccn.org/professionals/physician_gls/pdf/cml.pdf. (accessed 8.01.2019).

  17. Hensley ML, Ford JM. Imatinib treatment: specific issues related to safety, fertility, and pregnancy. Semin Hematol. 2003;40(2 Suppl 2):21–5. doi: 10.1053/shem.2003.50038.

  18. Cortes JE, Abruzzese E, Chelysheva E, et al. The impact of dasatinib on pregnancy outcomes. Am J Hematol. 2015;90(12):1111–5. doi: 10.1002/ajh.24186.

  19. Pye SM, Cortes J, Ault P, et al. The effects of imatinib on pregnancy outcome. Blood. 2008;111(12):5505–8. doi: 10.1182/blood-2007-10-114900.

  20. Berveiller P, Andreoli A, Mir O, et al. A dramatic fetal outcome following transplacental transfer of dasatinib. Anti-Cancer Drugs. 2012;23(7):754–7. doi: 10.1097/CAD.0b013e328352a8fe.

  21. Polin RA, Fox WW, Abman SH. Fetal and Neonatal Physiology. 4th edition; 2011. 2208 p. [Internet] Available from: http://readli.net/fetal-and-neonatal-physiology. (accessed 8.01.2019).

  22. Abruzzese E, Scortechini AR, Gugliotta G, et al. Gimema Registry of conception/pregnancy in adult patients diagnosed with chronic myeloid leukemia (CML) treated with tyrosine kinase inhibitors (TKIs). Blood. 2014;124:1806, abstract.

  23. Chelysheva E, Galaiko MV, Kolosheinova TI, et al. Outcomes of pregnancy and therapeutic approaches in chronic myeloid leukemia during pregnancy. Hematologica. 2014;99(Suppl 1):336–7.

  24. Jovelet C, Seck A, Mir O, et al. Variation in transplacental transfer of tyrosine kinase inhibitors in the human perfused cotyledon model. Ann Oncol. 2015;26(7):1500–4. doi: 10.1093/annonc/mdv172.

  25. Chelysheva E, Turkina A, Polushkina E, et al. Placental transfer of tyrosine kinase inhibitors used for chronic myeloid leukemia treatment. Leuk Lymphoma. 2018;59(3):733–8. doi: 10.1080/10428194.2017.1347929.

  26. Russel MA, Carpenter MW, Akhtar MS, et al. Imatinib mesylate and metabolite concentration in maternal blood, umbilical cord blood, placenta and breast milk. J Perinatol. 2007;27(4):241–3. doi: 10.1038/sj.jp.7211665.

  27. Cole S, Kantarjian H, Ault P, et al. Successful completion of pregnancy in a patient with chronic myeloid leukemia without active intervention: a case report and review of the literature. Clin Lymph Myel. 2009;9(4):324–7. doi: 10.3816/CLM.2009.n.064.

  28. Iqbal J, Ali Z, Khan AU, et al. Pregnancy outcomes in patients with chronic myeloid leukemia treated with imatinib mesylate: short report from a developing country. Leuk Lymphoma. 2014;55(9):2109–13. doi: 10.3109/10428194.2013.866662.

  29. Alizadeh H, Jaafar H, Rajnics P, et al. Outcome of pregnancy in chronic myeloid leukaemia patients treated with tyrosine kinase inhibitors: short report from a single centre. Leuk Res. 2015;39(1):47–51. doi: 10.1016/j.leukres.2014.10.002.

  30. Saussele S, Richter J, Hochhaus A, et al. The concept of treatment-free remission in chronic myeloid leukemia. Leukemia. 2016;30(8):1638–47. doi: 10.1038/leu.2016.115.

  31. Mahon FX, Rea D, Guilhot J, et al. Discontinuation of imatinib in patients with chronic myeloid leukaemia who have maintained complete molecular remission for at least 2 years: the prospective, multicentre Stop Imatinib (STIM) trial. Lancet Oncol. 2010;11(11):1029–35. doi: 10.1016/S1470-2045(10)70233-3.

  32. Rea D, Nicolini F, Tulliez M, et al. Discontinuation of dasatinib or nilotinib in chronic myeloid leukemia: interim analysis of the STOP 2G-TKI Study. Blood. 2017;129(7):846–54. doi: 10.1182/blood-2016-09-742205.

  33. Saussele S, Richter J, Guilhot J, et al Discontinuation of tyrosine kinase inhibitor therapy in chronic myeloid leukaemia (EURO-SKI): a prespecified interim analysis of a prospective, multicentre, non-randomised, trial. Lancet Oncol. 2018;19(6):747–7. doi: 10.1016/S1470-2045(18)30192-X.

  34. Rousselot P, Charbonnier A, Cony-Makhoul P, et al. Loss of major molecular response as a trigger for restarting tyrosine kinase inhibitor therapy in patients with chronic-phase chronic myelogenous leukemia who have stopped imatinib after durable undetectable disease. J Clin Oncol. 2014;32(5):424–30. doi: 10.1200/JCO.2012.48.5797.

  35. Chelysheva E, Apperley J, Abruzzese E, et al. Kinetics of the leukemic clone in patients with chronic myeloid leukemia during pregnancy. 2018;132(Suppl 1):4254, abstract.

  36. Burchert A, Muller MC, Kostrewa P, et al. Sustained molecular response with interferon alfa maintenance after induction therapy with imatinib plus interferon alfa in patients with chronic myeloid leukemia. J Clin Oncol. 2010;28(8):1429–35. doi: 10.1200/JCO.2009.25.5075.

  37. Law AD, Kim DHD, Lipton JH. Pregnancy: part of life in chronic myelogenous leukemia. Leuk Lymphoma. 2017;58(2):280–7. doi: 10.1080/10428194.2016.1201571.

  38. Patel M, Dukes IA, Hull JC. Use of hydroxyurea in chronic myeloid leukemia during pregnancy; a case report. Am J Obstet Gynecol. 1991;165(3):565–6. doi: 10.1016/0002-9378(91)90285-y.

  39. Tretian G, Tchernia G, Papiernik E, et al. Hydroxyurea and pregnancy. Am J Obstet Gynecol. 1992;166(6):1868. doi: 10.1016/0002-9378(92)91590-7.

  40. Assi R, Kantarjian HM, Keating MJ, et al. Management of chronic myeloid leukemia (CML) during pregnancy among patients (pts) treated with a tyrosine kinase inhibitor (TKI): a single-center experience. Blood. 2017;130:2881, abstract.

  41. Ali R, Ozkalemkas F, Kimya Y, et al. Imatinib use during pregnancy and breast feeding: a case report and review of the literature. Arch Gynecol Obstet. 2009;280(2):169–75. doi: 10.1007/s00404-008-0861-7.

  42. Chelysheva E, Aleshin S, Polushkina E, et al. Breastfeeding in patients with chronic myeloid leukaemia: case series with measurements of drug concentrations in maternal milk and literature review. Mediterr J Hematol Infect Dis. 2018;10(1):2018027. doi: 10.4084/MJHID.2018.027.

  43. Chelysheva E, Turkina A, Polushkina E, et al. Results of treatment of patients with chronic myeloid leukemia and pregnancy in accordance with the leukemic burden and term of pregnancy (the LET scheme). EHA Learning Center. 2018. Abstract PF375. Available from: https://learningcenter.ehaweb.org/eha/2018/stockholm/214848/ekaterina.chelysheva.results.of.treatment.of.patients.with.chronic.myeloid.html?f=menu=6*ce_id=1346*ot_id=19052*media=3*marker=167 (accessed 8.01.2019).

  44. Chelysheva E, Turkina A, Polushkina E, et al. Treatment of patients with chronic myeloid leukemia during pregnancy according to scheme considering the leukemic burden and term of pregnancy (the LRT scheme). Clin Lymph Myel Leuk. 2018;18:S227–8. doi: 10.1016/j.clml.2018.07.102.

  45. Chelysheva E, Abruzzese E, Rea D, et al. Chronic myeloid leukemia diagnosed during pregnancy: therapy, outcomes and follow-up. 2018;132(Suppl 1):4255, abstract.

  46. Abruzzese E, de Fabritiis P, Trawinska MM, et al. Back to the future: Treatment-free remission and pregnancy in chronic myeloid leukemia. Eur J Haematol. 2018;102(2):197–9. doi: 10.1111/ejh.13192.

Treatment of Chronic Myeloid Leukemia According to Current Guidelines: The Results of the Pilot Prospective Study “Early Induction Therapy and Monitoring” (РИТМ)

OA Shukhov, AG Turkina, EYu Chelysheva, AV Bykova, AN Petrova, GA Gusarova, IS Nemchenko, AO Abdullaev, TN Obukhova, AB Sudarikov

National Medical Hematology Research Center, 4а Novyi Zykovskii pr-d, Moscow, Russian Federation, 125167

For correspondence: Oleg Aleksandrovich Shukhov, MD, PhD, 4а Novyi Zykovskii pr-d, Moscow, Russian Federation, 125167; e-mail: shuhov@list.ru

For citation: Shukhov OA, Turkina AG, Chelysheva EYu, et al. Treatment of Chronic Myeloid Leukemia According to Current Guidelines: The Results of the Pilot Prospective Study “Early Induction Therapy and Monitoring” (РИТМ). Clinical oncohematology 2019;12(2):194–201.

DOI: 10.21320/2500-2139-2019-12-2-194-201


ABSTRACT

Background. Current clinical guidelines on diagnosis and treatment of chronic myeloid leukemia (CML) define indications for substitution of first-line tyrosine kinase inhibitor (TKI) at therapy failure during different phases of disease progression.

Aim. To assess the efficacy of CML treatment with implementing the protocol of timely monitoring and switching to another TKI.

Materials & Methods. Patients were included into pilot prospective study РИТМ during 5 years. Data on 100 CML patients were analyzed. Therapy and monitoring were conducted according to the Federal clinical guidelines on CML diagnosis and therapy, 2013.

Results. Median follow-up after initiation of treatment was 46 months (range 12–74). Imatinib mesylate was administered as first-line therapy to 91 (91 %) patients, 9 (9 %) patients received 2nd generation TKI (TKI2). Therapy failure was registered in 31 (31 %) patients; 26 (84 %) of them were switched to TKI2. At the time of analysis 95 (95 %) patients were followed-up. Cumulative incidence of CML-associated mortality was 2 %. By the fifth year of follow-up cumulative probability of complete cytogenetic, major and deep molecular responses was 93 %, 88 % and 66 %, respectively.

Conclusion. CML treatment according to current guidelines yields the results comparable with those achieved by first-line TKI2 therapy. This approach reduces CML treatment costs and lowers the risk of TKI2-associated adverse events. Due to a high rate of deep molecular response the proportion of CML patients in remission without treatment can be increased in the future.

Keywords: chronic myeloid leukemia, monitoring, tyrosine kinase inhibitors, TKI switch.

Received: October 21, 2018

Accepted: February 4, 2019

Read in PDF 


REFERENCES

  1. O’Brien SG, Guilhot F, Larson RA, et al. Imatinib compared with interferon and low-dose cytarabine for newly diagnosed chronic-phase chronic myeloid leukemia. N Engl J Med. 2003;348(11):994–1004. doi: 10.1056/NEJMoa022457.

  2. Hochhaus A, Larson RA, Guilhot F, et al. Long-term outcomes of imatinib treatment for chronic myeloid leukemia. N Engl Med. 2017;376(10):917–27. doi: 10.1056/NEJMoa1609324.

  3. Bower H, Bjorkholm M, Dickman PW, et al. Life expectancy of patients with chronic myeloid leukemia approaches the life expectancy of the general population. J Clin Oncol. 2016;34(24):2851–7. doi: 10.1200/JCO.2015.66.2866.

  4. Hochhaus A, Saglio G, Hughes TP, et al. Long-term benefits and risks of frontline nilotinib vs imatinib for chronic myeloid leukemia in chronic phase: 5-year update of the randomized ENESTnd trial. Leukemia. 2016;30(5):1044–54. doi: 10.1038/leu.2016.5.

  5. Cortes JE, Saglio G, Kantarjian HM, et al. Final 5-year study results of DASISION: the dasatinib versus imatinib study in treatment-naive chronic myeloid leukemia patients trial. J Clin Oncol. 2016;34(20):2333–40. doi: 10.1200/JCO.2015.64.8899.

  6. Cortes JE, Kantarjian HM, Brummendorf TH, et al. Safety and efficacy of bosutinib (SKI-606) in chronic phase Philadelphia chromosome-positive chronic myeloid leukemia patients with resistance or intolerance to imatinib. Blood. 2011;118(17):4567–76. doi: 10.1182/blood-2011-05-355594.

  7. Cortes JE, Kim DW, Pinilla-Ibarz J, et al. Ponatinib efficacy and safety in Philadelphia chromosome-positive leukemia: final 5-year results of the phase 2 PACE trial. Blood. 2018;132(4):393–404. doi: 10.1182/blood-2016-09-739086.

  8. Marin D, Ibrahim AR, Lucas C, et al. Assessment of BCR-ABL1 transcript levels at 3 months is the only requirement for predicting outcome for patients with chronic myeloid leukemia treated with tyrosine kinase inhibitors. J Clin Oncol. 2012;30(3):232–8. doi: 10.1200/JCO.2011.38.6565.

  9. Jabbour E, Kantarjian HM, Saglio G, et al. Early response with dasatinib or imatinib in chronic myeloid leukemia: 3-year follow-up from a randomized phase 3 trial (DASISION). Blood. 2014;123(4):494–500. doi: 10.1182/blood-2013-06-511592.

  10. Saglio G, Snedecor SJ, Xiang J, et al. Early molecular response (EMR) with frontline treatment is a significant predictor of long-term BCR-ABL transcript levels in patients with Philadelphia chromosome-positive chronic myeloid leukemia (CML) in chronic phase. Blood. 2015;126(23):1579,

  11. Dulucq S, Mahon FX. Deep molecular responses for treatment-free remission in chronic myeloid leukemia. Cancer Med. 2016;5(9):2398–411. doi: 10.1002/cam4.801.

  12. Baccarani M, Deininger MW, Rosti G, et al. European LeukemiaNet recommendations for the management of chronic myeloid leukemia: 2013. Blood. 2013;122(6):872–84. doi: 10.1182/blood-2013-05-501569.

  13. National Comprehensive Cancer Network. Chronic myeloid leukemia. Version 1.2019. Available from: https://www.nccn.org/professionals/physician_gls/pdf/cml.pdf. (accessed 3.12.2018).

  14. Абдулкадыров К.М., Абдуллаев А.О., Авдеева Л.Б. и др. Федеральные клинические рекомендации по диагностике и терапии хронического миелолейкоза. Вестник гематологии. 2013;9(3):4–41.

    [Abdulkadyrov KM, Abdullaev AO, Avdeeva LB, et al. Federal clinical guidelines for diagnosis and treatment of chronic myeloid leukemia. Vestnik gematologii. 2013;9(3):4–41. (In Russ)]

  15. Туркина А.Г., Зарицкий А.Ю., Шуваев В.А. и др. Клинические рекомендации по диагностике и лечению хронического миелолейкоза. Клиническая онкогематология. 2017;10(3):294–316. doi: 21320/2500-2139-2017-10-3-294-316.

    [Turkina AG, Zaritskii AYu, Shuvaev VA, et al. Clinical Recommendations for the Diagnosis and Treatment of Chronic Myeloid Leukemia. Clinical oncohematology. 2017;10(3):294–316. doi: 10.21320/2500-2139-2017-10-3-294-316. (In Russ)]

  16. Klil-Drori AJ, Yin H, Azoulay L, et al. Early switch to second-line tyrosine kinase inhibitor in chronic myeloid leukemia patients failing to achieve early molecular response. Am J Hematol. 2017;92(10):E602–4. doi: 10.1002/ajh.24838.

  17. Milojkovic D, Clarck RE, Byrne JL, et al. The Target UK study: Real-world evidence of molecular response to tyrosine kinase inhibitors supports European LeukemiaNet 2013 recommendations for the management of chronic myeloid leukaemia. Blood. 2017;130(1):2892, abstract.

  18. Yeung DT, Osborn MP, White DL, et al. TIDEL-II: first-use of imatinib in CML with early switch to nilotinib for failure to achieve time-dependent molecular targets. Blood. 2015;125(6):915–23. doi: 10.1182/blood-2014-07-590315.

  19. Mahon FX. Discontinuation of tyrosine kinase therapy in CML. Ann Hematol. 2015;94(Suppl 2):187–93. doi: 10.1007/s00277-015-2320-4.

  20. Hochhaus A, Masszi T, Giles FJ, et al. Treatment-free remission following frontline nilotinib in patients with chronic myeloid leukemia in chronic phase: results from the ENESTfreedom study. Leukemia. 2017;31(7):1525–31. doi: 10.1038/leu.2017.63.

  21. Mahon FX, Boquimpani C, Kim DW, et al. Treatment-free remission after second-line nilotinib treatment in patients with chronic myeloid leukemia in chronic phase: results from a single-group, phase 2, open-label study. Ann Intern Med. 2018;168(7):461–70. doi: 10.7326/M17-1094.

  22. Etienne G, Guilhot J, Rea D, et al. Long-Term Follow-Up of the French Stop Imatinib (STIM1) Study in Patients With Chronic Myeloid Leukemia. J Clin Oncol. 2017;35(3):298–305. doi: 10.1200/JCO.2016.68.2914.

  23. Saussele S, Richter J, Guilhot J, et al. Discontinuation of tyrosine kinase inhibitor therapy in chronic myeloid leukaemia (EURO-SKI): a prespecified interim analysis of a prospective, multicentre, non-randomised, trial. Lancet Oncol. 2018;19(6):747–57. doi: 10.1016/S1470-2045(18)30192-X.

  24. Hughes TP, Ross DM. Moving treatment-free remission into mainstream clinical practice in CML. Blood. 2016;128(1):17–23. doi: 10.1182/blood-2016-01-694265.

  25. Тасигна® (инструкция по медицинскому применению). Швейцария: Novartis pharma, AG. Доступно по: https://www.vidal.ru/drugs/tasigna__42603. Ссылка активна на12.2018.

    [Tasigna® (package insert). Switzerland: Novartis pharma, AG. Available from: https://www.vidal.ru/drugs/tasigna__42603. (accessed 3.12.2018) (In Russ)]

  26. Rea D, Nicolini FE, Tulliez M, et al. Discontinuation of dasatinib or nilotinib in chronic myeloid leukemia: interim analysis of the STOP 2G-TKI study. Blood. 2017;129(7):846–54. doi: 10.1182/blood-2016-09-742205.

  27. ТуркинаА.Г., Челышева Е.Ю., Шуваев В.А. и др. Результаты наблюдения больных хроническим миелолейкозом с глубоким молекулярным ответом без терапии ингибиторами тирозинкиназ. Терапевтический архив. 2017;89(12):86–96. doi: 10.17116/terarkh2017891286-96.

    [Turkina AG, Chelysheva EYu, Shuvaev VA, et al. Results of following up patients with chronic myeloid leukemia and a deep molecular response without tyrosine kinase inhibitor therapy. Terapevticheskii arkhiv. 2017;89(12):86–96. doi: 10.17116/terarkh2017891286-96. (In Russ)]

  28. Туркина А.Г., Челышева Е.Ю. Стратегия терапии хронического миелолейкоза: возможности и перспективы. Терапевтический архив. 2013;85(7):4–9.

    [Turkina AG, Chelysheva EYu. Therapeutic strategy for chronic myeloid leukemia: possibilities and prospects. Terapevticheskii arkhiv. 2013;85(7):4–9. (In Russ)]

  29. Шуваев В.А., Абдулкадыров К.М., Туркина А.Г. и др. Фармакоэкономический анализ ремиссии хронического миелолейкоза без лечения. Гематология и трансфузиология. 2015;60(4):14–20.

    [Shuvaev VA, Abdulkadyrov KM, Turkina AG, et al. Pharmacoeconomic analysis of chronic myeloid leukemia remission without treatment. Gematologiya i transfuziologiya. 2015;60(4):14–20. (In Russ)]

Expression of the BCR-ABL1 Gene in Patients with Chronic Myeloproliferative Diseases with Signs of Progression

LA Kesaeva1, EN Misyurina2, DS Mar’in2, EI Zhelnova2, AYu Bulanov2, AE Misyurina3, AA Krutov4, IN Soldatova4, SS Zborovskii4, VA Misyurin1,4, VV Tikhonova1, YuP Finashutina1, ON Solopova1, NA Lyzhko1, AE Bespalova1, NN Kasatkina1, AV Ponomarev1, MA Lysenko2, AV Misyurin1,4

1 NN Blokhin National Medical Cancer Research Center, 24 Kashirskoye sh., Moscow, Russian Federation, 115478

2 Municipal Clinical Hospital No. 52, 3 Pekhotnaya str., Moscow, Russian Federation, 123182

3 National Research Center for Hematology, 4 Novyi Zykovskii pr-d, Moscow, Russian Federation, 125167

4 GenoTekhnologiya, 104 Profsoyuznaya str., Moscow, Russian Federation, 117485

For correspondence: Andrei Vital’evich Misyurin, PhD in Biology, 24 Kashirskoye sh., Moscow, Russian Federation, 115478; Tel.: +7(499)612-80-38; e-mail: and@genetechnology.ru

For citation: Kesaeva LA, Misyurina EN, Mar’in DS, et al. Expression of the BCR-ABL1 Gene in Patients with Chronic Myeloproliferative Diseases with Signs of Progression. Clinical oncohematology. 2018;11(4):354–9.

DOI: 10.21320/2500-2139-2018-11-4-354-359


ABSTRACT

Background. The V617F mutation of JAK2 is known to manifest in Ph-negative chronic myeloproliferative diseases (cMPD), such as polycythemia vera, thrombocythemia, and myelofibrosis. These diseases not infrequently advance into more aggressive forms up to acute leukemia. As the progression mechanism is still unknown, its study retains a high priority. JAK2 carrying the V617F mutation is believed to cause constant activation of V(D)J recombinase in myeloid tumor cells in cMPD patients. Aberrant activation of V(D)J recombinase in tumor cells in cMPD patients can lead to t(9;22)(q34;q11) chromosomal rearrangement.

Aim. To study the expression of BCR-ABL1 resulting from translocation t(9;22)(q34;q11) in cMPD patients at the progression stage in order to test the suggested hypothesis.

Materials & Methods. The BCRABL1 expression was assessed in peripheral blood granulocytes in cMPD patients by real-time PCR. The JAK2 V617F mutation was identified by quantitative allele-specific PCR. The JAK2 exon 12 mutations were determined using Sanger direct sequencing of PCR products.

Results. The BCR-ABL1 expression was discovered in 29 % of patients with cMPD progression. The BCR-ABL1 expression in these patients correlated with hepatosplenomegaly and hyperleukocytosis.

Conclusion. In a significant proportion of cMPD patients the disease progression can be associated with activation of the BCR-ABL expression.

Keywords: JAK2 V617F, BCR-ABL1, V(D)J recombinase, t(9;22)(q34;q11), polycythemia vera, essential thrombocythemia, myelofibrosis, chronic myeloid leukemia.

Received: April 2, 2018

Accepted: August 5, 2018

Read in PDF 


REFERENCES

  1. Dameshek W. Some speculations on the myeloproliferative syndromes. Blood. 1951;6(4):372–5.

  2. Nowell P, Hungerford D. A minute chromosome in human chronic granulocytic leukemia. Science. 1960;132:1497, abstract.

  3. Rowley JD. Letter: A new consistent chromosomal abnormality in chronic myelogenous leukemia identified by quinacrine fluorescence and Giemsa staining. Nature. 1973;243(5405):290–3. doi: 10.1038/243290a0.

  4. Davis R, Konopka J, Witte O. Activation of the c-abl оncogene by viral transduction or chromosomal translocation generates altered c-abl proteins with similar in vitro kinase properties. Mol Cell Biol. 1985;5(1):204–13. doi: 10.1128/mcb.5.1.204.

  5. Muller AJ, Young JC, Pendergast AM, et al. BCR first exon sequences specifically activate the BCR/ABL thyrosine kinase oncogene of Philadelphia chromosome-positive human leukemias. Mol Cell Biol. 1991;11(4):1785–92. doi: 10.1128/mcb.11.4.1785.

  6. James C, Ugo V, Le Couedic JP, et al. A unique clonal JAK2 mutation leading to constitutive signaling causes polycythaemia vera. Nature. 2005;434(7037):1144–8. doi: 10.1038/nature03546.

  7. Scott LM, Tong W, Levine RL, et al. JAK2 exon 12 mutations in polycythemia vera and idiopathic erythrocytosis. N Engl J Med. 2007;356(5):459–68. doi: 10.1056/NEJMoa065202.

  8. Pikman Y, Lee BH, Mercher T, et al. MPLW515L is a novel somatic activating mutation in myelofibrosis with myeloid metaplasia. PLoS Med. 2006;3(7):e270. doi: 10.1371/journal.pmed.0030270.

  9. Klampfl T, Gisslinger H, Harutyunyan AS, et al. Somatic mutations of calreticulin in myeloproliferative neoplasms. N Engl J Med. 2013;369(25):2379–90. doi: 10.1056/NEJMoa1311347.

  10. Nangalia J, Massie CE, Baxter EJ, et al. Somatic CALR mutations in myeloproliferative neoplasms with nonmutated JAK2. N Engl J Med. 2013;369(25):2391–405. doi: 10.1056/NEJMoa1312542.

  11. Tutaeva V, Misurin AV, Rozenberg JM, et al. Application of PRV-1 mRNA expression level and JAK2V617F mutation for the differentiating between polycytemia vera and secondary erythrocytosis and assessment of treatment by interferon or hydroxyurea. Hematology. 2007;12(6):473–9. doi: 10.1080/10245330701384005.

  12. Мисюрин А.В. Молекулярный патогенез миелопролиферативных заболеваний. Клиническая онкогематология. 2009;2(3):211–9.

    [Misyurin AV. Molecular pathogenesis of myeloproliferative disorders. Klinicheskaya onkogematologiya. 2009;2(3):211–9. (In Russ)]

  13. Vainchenker W, Delhommeau F, Constantinescu SN, Bernard OA. New mutations and pathogenesis of myeloproliferative neoplasms. Blood. 2011;118(7):1723–35. doi: 10.1182/blood-2011-02-292102.

  14. Mirza I, Frantz C, Clarce G, et al. Transformation of polycythemia vera to chronic myelogenous leukemia. Arch Pathol Lab Med. 2007;131(11):1719–24.

  15. Toogeh G, Ferdowsi S, Naadali F, et al. Concomitant presence of JAK2 V617F mutation and BCR-ABL translocation in a pregnant woman with polycythemia vera. Med Oncol. 2011;28(4):1555–8. doi: 10.1007/s12032-010-9570-8.

  16. Bee PC, Gan GG, Nadarajan VS, et al. A man with concomitant polycythaemia vera and chronic myeloid leukemia: the dynamics of the two disorders. Int J Hematol. 2010;91(1):136–9. doi: 10.1007/s12185-009-0471-6.

  17. Kemp NH, Stafford JL, Tanner R. Chromosome studies during early and terminal chronic myeloid leukemia. Br Med J. 1964;1(5389):1010–4. doi: 10.1136/bmj.1.5389.1010.

  18. Hoppin EC, Lewis JP. Polycythemia Rubra Vera Progressing to Ph-Positive Chronic Myelogenous Leukemia. Ann Intern Med. 1975;83(6):820–3. doi: 10.7326/0003-4819-83-6-820.

  19. Saviola A, Claudia Fiorani C, Ferrara L. Transition of polycythemia vera to chronic myeloid leukaemia. Eur J Haematol. 2005;75(3):264–6. doi: 10.1111/j.1600-0609.2005.00488.x.

  20. Мисюрин А.В., Сурин В.Л., Тагиев А.Ф. Новые точки разрыва транслокации t(9;22) при хроническом миелолейкозе. Биоорганическая химия. 1999;25(3):234–6.

    [Misyurin AV, Surin VL, Tagiev AF. New breakpoints of translocation t(9;22) in chronic myeloid leukemia. Bioorganicheskaya khimiya. 1999;25(3):234–6. (In Russ)]

  21. Score J, Calasanz MJ, Ottman O, et al. Analysis of genomic breakpoints in p190 and p210 BCR-ABL indicate distinct mechanisms of formation. Leukemia. 2010;24(10):1742–50. doi: 10.1038/leu.2010.174.

  22. Bassing CH, Swat W, Alt FW. The mechanism and regulation of chromosomal V(D)J recombination. Cell. 2002;109(2):S45–S55. doi: 10.1016/S0092-8674(02)00675-X.

Epidemiology of Chronic Myeloid Leukemia in the Republic of Bashkortostan

NR Ryabchikova, GSh Safuanova, VI Nikulicheva

Bashkir State Medical University, 3 Lenina str., Ufa, Russian Federation, 450008

For correspondence: Prof. Guzyal’ Shagbanovna Safuanova, MD, PhD, 3 Lenina str., Ufa, Russian Federation, 450008; Tel.: +7(927)639-03-73; e-mail: safuanova@bk.ru

For citation: Ryabchikova NR, Safuanova GSh, Nikulicheva VI. Epidemiology of Chronic Myeloid Leukemia in the Republic of Bashkortostan. Clinical oncohematology. 2018;11(4):349–53.

DOI: 10.21320/2500-2139-2018-11-4-349-353


ABSTRACT

Background. The planning of therapeutic, diagnostic, and preventive medical care for chronic myeloid leukemia (CML) patients implies the need of not only maintaining patient registries, but also conducting epidemiologic studies in each geographical area.

Aim. To study and analyze CML epidemiological indicators over the last 15 years in the Republic of Bashkortostan for the purposes of evaluation and rational planning of specialized medical care for CML patient population.

Materials & Methods. The incidence, prevalence, and mortality of CML patients of all age groups in the period of 2000–2016 was analyzed in the Republic of Bashkortostan.

Results. The analysis of epidemiological indicators over the period of 2000–2016 showed that the incidence of the disease in the Republic of Bashkortostan was increasing. Within the last 8 years the prevalence rate even quadrupled which is clearly connected with improved detectability of Ph-chromosome and/or BCR-ABL gene, creation and maintenance of CML patient registry since 2008, introduction of treatment using tyrosine kinase inhibitors resulting also in increase in life expectancy. Mortality rates are reported to have a tendency of decrease over the period under study.

Conclusion. Key epidemiological indicators of CML in the Republic of Bashkortostan are comparable with the data of international and Russian researchers. The results obtained can be used for comparative studies and improvement of specialized medical care for CML patients.

Keywords: chronic myeloid leukemia, epidemiology, incidence, prevalence, mortality.

Received: April 9, 2018

Accepted: August 3, 2018

Read in PDF 


REFERENCES

  1. Абдулкадыров К.М., Абдуллаев А.О., Авдеев Л.Б. и др. Федеральные клинические рекомендации по диагностике и терапии хронического миелолейкоза. Вестник гематологии. 2013;9(3):4–40.

    [Abdulkadyrov KM, Abdullaev AO, Avdeev LB, et al. Fedеral clinical guidelines on diagnosis and treatment of chronic myeloid leukemia. Vestnik gematologii. 2013;9(3):4–40. (In Russ)]

  2. Волкова С.А., Ковалишена О.В., Прыткова М.В. и др. Эпидемиологическое исследование хронического миелолейкоза у взрослого населения Нижегородской области за период 1980–2003 гг. Гематология и трансфузиология. 2005;50(2):8–13.

    [Volkova SA, Kovalishena OV, Prytkova MV, et al. Epidemiological studies of chronic myeloid leukemia in adults of the Nizhny Novgorod region in 1980–2003. Gematologiya i transfuziologiya. 2005;50(2):8–13. (In Russ)]

  3. Виноградова О.Ю., Куликов С.М., Куцев С.М. и др. Проблемы организации лечения хронического миелолейкоза в России. Клиническая онкогематология. 2011;4(4):292–7.

    [Vinogradova OYu, Kulikov SM, Kutsev SM, et al. Issues of organizing the treatment of chronic myeloid leukemia in the Russian Federation. Klinicheskaya onkogematologiya. 2011;4(4):292–7. (In Russ)]

  4. Pasguini R, Cortes J, Kantarjian HM, et al. A worldwide observational registry collecting longitudinal data on management of chronic myeloid leukemia patients (The WORLD CML Registry)-2nd Annual interim analysis. Blood. 2010;116(21):2292.

  5. Juliusson G, Lazarevic V, Horsterdt A, et al. Acute myeloid leukemia in the real world: why population-based registries are needed. Blood. 2012;119(17):3890–9. doi: 10.1182/blood-2011-12-379008.

  6. Варшавский А.В. Клинико-эпидемиологическая характеристика гемобластозов в Республике Башкортостан: Автореф. дис.… канд. мед. наук. Уфа, 2011.

    [Varshavskii AV. Kliniko-epidemiologicheskaya kharakteristika gemoblastozov v Respublike Bashkortostan. (Clinical and epidemiological characteristics of hemoblastosis in the Republic of Bashkortostan.) [dissertation] Ufa; 2011. (In Russ)]

  7. Рябчикова Н.Р., Минниахметов И.Р, Сафуанова Г.Ш. и др. Хронический миелолейкоз: молекулярный мониторинг в клинической практике. Онкогематология. 2013;8(1):1–16. doi: 10.17650/1818-8346-2013-8-1-7-16.

    [Ryabchikova NR, Minniakhmetov IR, Safuanova GSh, et al. Chronic myeloid leukemia: molecular monitoring in clinical practice. Onkogematologiya. 2013;8(1):1–16. doi: 10.17650/1818-8346-2013-8-1-7-16. (In Russ)]

  8. Куликов С.М., Виноградова О.Ю., Челышева Е.Ю. и др. Заболеваемость хроническим миелолейкозом в 6 регионах России по данным популяционного исследования 2009–2012 гг. Терапевтический архив. 2014;86(7):24–30.

    [Kulikov SM, Vinogradova OYu, Chelysheva EYu, et al. Incidence of chronic myeloid leukemia in 6 regions of Russia according to the data of the 2009–2012 population-based study. Terapevticheskii arkhiv. 2014;86(7):24–30. (In Russ)]

  9. Туркина А.Г., Голенков А.К., Напсо Л.И. и др. Российский регистр по лечению хронического миелоидного лейкоза в рутинной клинической практике: итоги многолетней работы. Эффективная фармакотерапия. 2015;10(1):8–13.

    [Turkina AG, Golenkov AK, Napso LI, et al. The Russian registry of chronic myeloid leukemia treatment in routine clinical practice: results of the long-term work. Effektivnaya farmakoterapiya. 2015;(10):8–13. (In Russ)]

  10. Волкова С.А., Ковалишена О.В., Гостюжова Е.А. и др. Эффект от терапии иматинибом по данным клинико-эпидемиологического мониторинга хронического миелолейкоза в Нижегородской области за период 2000–2010 г. Гематология и трансфузиология. 2011;56(4):17–22.

    [Volkova SA, Kovalishena OV, Gostyuzhova EA, et al. Imatinib therapy effect according to the results of clinical and epidemiological monitoring of chronic myeloid leukemia in the Nizhny Novgorod region over the period of 2000–2010. Gematologiya i transfuziologiya. 2011;56(4):17–22. (In Russ)]

Tyrosine Kinase Inhibitor Resistance in Patients with Chronic Myeloid Leukemia: A 10-Year Study of BCR-ABL Gene Mutation Profile in Russia (2006–2016)

VV Tikhonova1,2, MA Isakov3, VA Misyurin1, YuP Finashutina1,2, LA Kesaeva1,2, NA Lyzhko 1, IN Soldatova2, NN Kasatkina1, EN Misyurina4, AV Misyurin1,2

1 NN Blokhin National Medical Cancer Research Center, 24 Kashirskoye sh., Moscow, Russian Federation, 115478

2 GenoTekhnologiya, 104 Profsoyuznaya str., Moscow, Russian Federation, 117485

3 Aston Consulting, 31g Shabolovka str., Moscow, Russian Federation, 115162

4 Municipal Clinical Hospital No. 52, 3 Pekhotnaya str., Moscow, Russian Federation, 123182

For correspondence: Vera Vyacheslavovna Tikhonova, 24 Kashirskoye sh., Moscow, Russian Federation, 115478; Tel.: +7(967)008-02-84; e-mail: brilfor@mail.ru

For citation: Tikhonova VV, Isakov MA, Misyurin VA, et al. Tyrosine Kinase Inhibitor Resistance in Patients with Chronic Myeloid Leukemia: A 10-Year Study of BCR-ABL Gene Mutation Profile in Russia (2006–2016). Clinical oncohematology. 2018;11(3):227–33.

DOI: 10.21320/2500-2139-2018-11-3-227-233


ABSTRACT

Background. Kinase domain mutations of BCR-ABL gene is the most common cause of tyrosine kinase inhibitor resistance.

Aim. To present the data on prognostic value of BCR-ABL mutation burden in Russian patients over the last 10 years.

Materials & Methods. The study included 1885 chronic myeloid leukemia (CML) patients with tyrosine kinase inhibitor resistance who were followed up from 2006 to 2016. BCR-ABL point mutations in mRNA samples were analyzed by means of polymerase chain reaction and subsequent Sanger sequencing.

Results. In 1257 CML patients with signs of tyrosine kinase inhibitor resistance BCR-ABL expression level was > 1 %. BCRABL mutations were detected in 31.8 % of patients. Total mutation count was 467 (70 mutation types). Total count of patients with mutation-associated tyrosine kinase inhibitor resistance decreased from 36.6 % (2006–2008) to 24.95 % (2013–2016) and to marked decrease of 23.12 % in 2014. Detection rate of imatinib-resistant mutations and F359V mutation was shown to decrease within the period from 2010–2011 to 2014–2015. F317L level, which is responsible for dasatinib resistance, considerably increased in 2015. T315I frequency was the highest in 2014, afterwards it was gradually decreasing. Mutation-associated resistance rates varied by region of the Russian Federation.

Conclusion. The analysis of trends of mutation incidence in patients with CML can be of extreme significance in long-term prognosis of resistance development and in improvement of treatment planning.

Keywords: chronic myeloid leukemia, kinase domain mutations of BCR-ABL gene, targeted therapy, resistance.

Received: January 22, 2018

Accepted: April 16, 2018

Read in PDF 


REFERENCES

  1. Soverini S, Colarossi S, Gnani A, et al. Contribution of ABL kinase domain mutations to imatinib resistance in different subsets of Philadelphia-positive patients: by the GIMEMA Working Party on Chronic Myeloid Leukemia. Clin Cancer Res. 2006;12(24):7374–9. doi: 10.1158/1078-0432.ccr-06-1516.
  2. Baccarani M, Cortes J, Pane F, et al. Chronic myeloid leukemia: an update of concepts and management recommendations of European Leukemia Net. J Clin Oncol. 2009;27(35):6041–51. doi: 10.1200/JCO.2009.25.0779.
  3. Soverini S, Rosti G, Iacobucci I, et al. Choosing the best second-line tyrosine kinase inhibitor in imatinib-resistant chronic myeloid leukemia patients harboring Bcr-Abl kinase domain mutations: how reliable is the IC50? Oncologist. 2011;16(6):868–76. doi: 10.1634/theoncologist.2010-0388.
  4. Овсянникова Е.Г., Капланов К.Д., Клиточенко Т.Ю. и др. Мутационный статус резистентных к иматинибу больных хроническим миелолейкозом. Онкогематология. 2012;4:16–24.[Ovsyannikova EG, Kaplanov KD, Klitochenko TYu, et al. Mutation status of chronic myeloid leukemia patients with imatinib resistance. Onkogematologiya. 2012;4:16–24. (In Russ)]
  5. Patkar N, Ghodke K, Joshi S, et al. Characteristics of BCR-ABL kinase domain mutations in chronic myeloid leukemia from India: not just missense mutations but insertions and deletions are also associated with TKI resistance. Leuk Lymphoma. 2016;57(11):2653–60. doi: 10.3109/10428194.2016.1157868.
  6. Elnahass YH, Mahmoud HK, Ali FT, et al. Abl Kinase Domain Mutations in Imatinib-treated Egyptian Patients with Chronic Myeloid Leukemia. J Leuk. 2013;1(1):106. doi: 10.4172/2329-6917.1000106.
  7. Awidi A, Ababneh N, Magablah A, et al. ABL Kinase Domain Mutations in Patients with Chronic Myeloid Leukemia in Jordan. Genet Test Mol Biomark. 2012;16(11):1317–21. doi: 10.1089/gtmb.2012.0147.
  8. Elias MH, Baba AA, Husin A, et al. Contribution of BCR-ABL kinase domain mutations to imatinib mesylate resistance in Philadelphia chromosome positive Malaysian chronic myeloid leukemia patients. Hematol Rep. 2012;4(4):e23. doi: 10.4081/hr.2012.e23.
  9. Vaidya S, Vundinti BR, Shanmukhaiah C, et al. Evolution of BCR/ABL Gene Mutation in CML Is Time Dependent and Dependent on the Pressure Exerted by Tyrosine Kinase Inhibitor. PLoS One. 2015;10(1):e0114828. doi: 10.1371/journal.pone.0114828.
  10. Челышева Е.Ю., Шухов О.А., Лазарева О.В., Туркина А.Г. Мутации киназного домена гена BCR-ABL при хроническом миелолейкозе. Клиническая онкогематология. 2012;5(1):13–21.[Chelysheva EYu, Shukhov OA, Lazareva OV, Turkina AG. Kinase domain mutations of BCR-ABL gene in patients with chronic myeloid leukemia. Klinicheskaya onkogematologiya. 2012;5(1):13–21. (In Russ)]
  11. Kimura S, Ando T, Kojima K. BCR-ABL Point Mutations and TKI Treatment in CML Patients. J Hematol Transfus. 2014;2(3):1022.
  12. Soverini S, de Benedittis C, Mancini M, Martinelli G. Mutations in the BCR-ABL1 Kinase Domain and Elsewhere in Chronic Myeloid Leukemia. Clin Lymph Myel Leuk. 2015;15(Suppl):S120–8. doi: 10.1016/j.clml.2015.02.035.
  13. Soverini S, De Benedittis C, Papayannidis C, et al. Drug resistance and BCR-ABL kinase domain mutations in Philadelphia chromosome-positive acute lymphoblastic leukemia from the imatinib to the second-generation tyrosine kinase inhibitor era: The main changes are in the type of mutations, but not in the frequency of mutation involvement. 2014;120(7):1002–9. doi: 10.1002/cncr.28522.
  14. Мисюрин А.В., Мисюрина Е.Н., Тихонова В.В. и др. Частота встречаемости мутаций киназного домена гена BCR-ABL у больных хроническим миелолейкозом, резистентных к терапии иматинибом. Российский биотерапевтический журнал. 2016;15(4):102–9. doi: 10.17650/1726-9784-2016-15-4-102-109.[Misyurin AV, Misyurina EN, Tikhonova VV, et al. BCR-ABL gene kinase domain mutation frequency in imatinib resistant chronic myeloid leukemia patients. Rossiiskii bioterapevticheskii zhurnal. 2016;15(4):102–9. doi: 10.17650/1726-9784-2016-15-4-102-109. (In Russ)]
  15. Hughes TP, Saglio G, Quintas-Cardama A, et al. BCR-ABL1 mutation development during first-line treatment with dasatinib or imatinib for chronic myeloid leukemia in chronic phase. Leukemia. 2015;29(9):1832–8. doi: 10.1038/leu.2015.168.
  16. Абдулкадыров К.М., Шуваев В.А., Фоминых М.С. Дженерики иматиниба: мифы и реальность (обзор литературы и собственные данные). Клиническая онкогематология. 2014;7(3):311–6.[Abdulkadyrov KM, Shuvaev VA, Fominykh MS. Imatinib Generics: Myths and Reality (Literature Review and Our Experience). Klinicheskaya onkogematologiya. 2014;7(3):311–6. (In Russ)]
  17. Валиев Т.Т., Левашов А.С., Сенжапова Э.Р. Таргетные препараты в детской онкологии. Онкопедиатрия. 2016;3(1):8–15. doi: 10.15690/onco.v3i1.1524.[Valiev TT, Levashov AS, Senzhapova ER. Targeted Drugs in Pediatric Oncology. Onkopediatriya. 2016;3(1):8–15. doi: 10.15690/onco.v3i1.1524. (In Russ)]